Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of...

51
V Biomedical Diagnostics II: Optical Biopsy

Transcript of Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of...

Page 1: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

VBiomedicalDiagnostics II: Optical Biopsy

Page 2: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

28Fluorescence

Spectroscopy forBiomedical Diagnostics

28.1 Introduction28.2 Principles of Fluorescence Spectroscopy

Fluorescence Techniques • Photophysical Basis of Luminescence

28.3 Characterization of LuminescenceEmission, Excitation, and Synchronous Spectra • Quantum Yields • Lifetimes • Polarization

28.4 Biomedical ApplicationsBiochemical Analysis of Individual Species • In Vitro Analyses and Diagnostics • In Vivo Analyses and Diagnostics

28.5 ConclusionAcknowledgmentsReferences

28.1 Introduction

In the past several decades, fluorescence spectroscopy has had a dramatic effect on many different fieldsof research. One field that has seen significant advancements is biomedical diagnostics. Within this field,fluorescence spectroscopy has been applied to the analysis of many different types of samples, rangingfrom individual biochemical species (e.g., NADH, tryptophan) to organs of living people. These studieshave given rise to new methods for the early or noninvasive diagnosis of various medical conditions,including tooth decay, atherosclerosis, heart arrhythmia, cancer, and many others.*

The medical condition that has seen the largest effort toward fluorescence-based analyses and trulydemonstrates the potential of fluorescence-based diagnoses is cancer. Fluorescence spectroscopy andimaging have been investigated for the diagnosis of almost every type of cancer and early neoplasticdifference found in humans. The detection of early neoplastic changes is important from an outcomeviewpoint because, once invasive carcinoma and metastases have occurred, treatment is difficult. Atpresent, excisional biopsy followed by histology is considered the “gold standard” for diagnosis of earlyneoplastic changes and carcinoma. In some cases, cytology rather than excisional biopsy is performed.These techniques are powerful diagnostic tools because they provide high-resolution spatial and mor-phological information of the cellular and subcellular structures of tissues. The use of staining andprocessing can enhance contrast and specificity of histopathology. However, both of these diagnosticprocedures require physical removal of specimens, followed by tissue processing in a laboratory. These

*Current affiliation: University of Maryland Baltimore County, Baltimore, Maryland

Tuan Vo-DinhOak Ridge National LaboratoryOak Ridge, Tennessee

Brian M. Cullum*Oak Ridge National LaboratoryOak Ridge, Tennessee

©2003 CRC Press LLC

Page 3: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

procedures incur a relatively high cost because specimen handling is required; more importantly, diag-nostic information is not available in real time. Moreover, in the context of detecting early neoplasticchanges, excisional biopsy and cytology can have unacceptable false negative rates often arising fromsampling errors.

Fluorescence techniques have the potential for performing in vivo diagnosis on tissue without the needfor sample excision and processing. Another advantage of fluorescence-based diagnoses is that theresulting information can be available in real time. In addition, because removal of tissue is not requiredfor optical diagnoses, a more complete examination of the organ of interest can be achieved than withexcisional biopsy or cytology.

This chapter provides an overview of fluorescence spectroscopy and its basic principles as well as itsbiomedical applications. Because biomedical fluorescence spectroscopy represents an extremely large andgrowing field of research, we have attempted to classify the ongoing research and clinical studies intothree different categories: (1) biochemical analyses of individual compounds, (2) in vitro analyses (cellularand tissue systems), and (3) in vivo analyses (animal and human studies). In addition to the overviewof fluorescence spectroscopy in this chapter, special methods and instrumental aspects for fluorescenceanalyses are further described in other chapters in this handbook: Chapter 33 on near-infrared fluores-cence imaging and spectroscopy in random media and tissues, Chapter 9 on lifetime-based imaging,microscopy, Chapter 10 on confocal miscroscopy, and Chapter 11 on two-photon excitation fluorescencemicroscopy.

28.2 Principles of Fluorescence Spectroscopy

28.2.1 Fluorescence Techniques

Luminescence is a branch of spectroscopy that deals primarily with the electronic states of an atom ormolecule, as opposed to the vibrational, rotational, or nuclear energy states. Fluorescence and phospho-rescence are the two sister techniques covered by the umbrella known as “luminescence spectroscopy.”These techniques involve the optical detection and spectral analysis of light emitted by a substance under-going a transition from an excited electronic state to a lower electronic state. Most luminescence-basedmedical diagnoses involve fluorescence spectroscopy because organic compounds fluoresce and do notphosphoresce at room temperature. In most phosphorescence studies, the measurements need to beperformed using low-temperature frozen solvents to minimize collisional quenching. A unique techniquecalled room temperature phosphorescence (RTP) uses various solid substrates to adsorb analytes into rigidmatrices (e.g., cellulose, silica gel, and alumina) in order to allow phosphorescence measurements.1 Becausethe vast majority of medical applications involve fluorescence analysis, this chapter focuses mainly on thetheory of fluorescence. However, because phosphorescence is also involved in the field of photodynamictherapy (PDT), we will also describe the relationship between fluorescence and phosphorescence.

Fluorescence diagnostic methods can be grouped into two main categories:

1. Methods that detect endogenous fluorophores in tissues2. Methods that detect or use exogenous fluorophores or fluorophore precursors [such as 5-amino-

levulinic acid (ALA)]

Fluorescence that originates from native fluorescent chromophores already present in the tissue isoften referred to as “autofluorescence.” Fluorescence may also originate from administered exogenouschromophores that have been synthesized to target specific tissues (e.g., dysplastic vs. normal), or it maybe activated by functional changes in the tissue. In a fluorescence analysis, excitation light at some specificwavelength (typically near-ultraviolet or visible) excites the tissue or exogenous fluorophore moleculesand induces fluorescence emission. Then some measure of the fluorescent emission is obtained, typicallyan emission spectrum (fluorescence emission intensity vs. wavelength).

©2003 CRC Press LLC

Page 4: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

28.2.2 Photophysical Basis of Luminescence

Most organic molecules contain an even number of electrons. In the ground state, the electrons fill thevarious atomic orbitals with the lowest energies in pairs. By the Pauli exclusion principle, two electronsin one given orbital must have spins in opposite directions, and the total spin S must equal 0. For thisreason, the ground state has no net electron spin; this state is called a “singlet” state.

Excitation of a molecule usually results in the promotion of one electron from the highest occupiedorbital to a previously unoccupied orbital (also referred to as a “virtual” orbital). Whereas the groundstate is generally a singlet, the excited state can be a singlet or a triplet, depending on the final spin stateof the electron promoted to the higher orbital. For illustration, Figure 28.1 shows one ground state andtwo excited-state configurations. The spins of the two electrons in the unoccupied orbitals are no longerrestricted by the Pauli exclusion principle. The singlet state has two antiparallel spins, whereas the tripletstate has two parallel spins and a net spin, S, of 1. In the general case, the configuration is not so easilyvisualized by schematic diagrams.2

The nature of the orbitals involved in an electronic transition is an important factor in determiningthe luminescence characteristics of the molecule. Organic compounds that are known to be stronglyluminescent are molecules with extensive π electron systems. These compounds include aromatic mol-ecules and a few unsaturated aliphatics. For molecules with no hetero-atom, electronic transitionsgenerally involve promotion of an electron from a bonding π orbital to an antibonding π* orbital. Sucha transition is known as a π – π* transition, and the resulting electronic state is called the ππ* excitedstate. For conjugated systems with hetero-atoms (N, O, or S) in the conjugated system, or with substituentscontaining the N, O, or S atoms, an electronic state may result from the promotion of an electron froma bonding n orbital to an antibonding π* orbital; this electronic state is called an nπ* excited state.

28.2.2.1 Molecular Electronic Energies

In the study of luminescence, we are first concerned with determination of the electronic states betweenwhich luminescence and other related processes such as intersystem crossing, vibrational relaxation, andinternal conversion occur. The basic approach for studying luminescence behavior of molecules is tocalculate the stationary-state electronic energies by solving the time-independent Schrödinger equation.A suitable wave function, Ψ, chosen to describe the properties of various molecular orbitals (MO) involvedin the photophysical processes, is given by the Schrödinger equation

H(q,Q)Ψ(q,Q) = EΨ(q,Q), (28.1)

FIGURE 28.1 Schematic diagram of typical spin arrangements in molecular orbitals for the ground, excited singlet,and triplet states.

MO

LEC

ULA

R O

RB

ITA

LS

EN

ER

GY

SINGLET SINGLET TRIPLET

EXCITEDSTATES

GROUNDSTATE

©2003 CRC Press LLC

Page 5: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

where H is the Hamiltonian operator of the molecule, q is the electronic spatial coordinates, Q is thenuclear spatial coordinates, and E is the energy eigenvalue.

The exact solutions for the Schrödinger equation are impossible to calculate for any molecular systemexcept H+. Therefore, calculations of electronic energies for excited singlet and triplet states of largersystems usually involve some method of approximation. The most commonly used method is the MOmethod, which utilizes one-electron orbitals to form molecular orbitals. One of the simplest of theseMO-based methods for calculating, or rather estimating, molecular electronic energies is the Hückelmolecular orbital (HMO) method, which treats certain integrals as parameters and selects these param-eters such that calculated values are in agreement with physical observables. This approach has beenquite successful in describing bonding in aromatic and highly conjugated molecules.

Figure 28.2, known as the Jablonskii diagram, shows energy levels for an organic molecule. In thisfigure, S0 …, Sn and T1…,Tm represent the discrete electronic energy levels of a molecule. The electronicstate having the lowest energy, S0 , is known as the ground state. S0 …, Sn and T1 …,Tm are the excitedsinglet and triplet states, respectively. Each electronic state has its own set of vibrational levels. Super-imposed on each vibrational level is a series of closely spaced rotational levels not shown in Figure 28.2.

28.2.2.2 Population of the Excited Electronic States

The main photophysical processes involved in the population and deactivation of excited electronic states,both singlet and triplet, are shown in Figure 28.2. These processes — absorption (A), vibrational relax-ation (VR), internal conversion (IC), fluorescence (F), intersystem crossing (ISC), and phosphorescence(P) — and related photophysical processes are discussed next.

Absorption (A)At equilibrium, a group of molecules has a thermal distribution at the lowest vibrational and rotationallevels of the ground state, S0. When a molecule absorbs excitation energy, it is elevated from S0 to somevibrational level of one of the excited singlet states, Sn, in the manifold S1,…, Sn. As postulated by theFranck–Condon principle, the time for an electronic transition (on the order of 10–15 s) is shorter thanthe time for nuclear rearrangement (on the order of 10–13 to 10–14 s). As a consequence, the molecularconfiguration following absorption is often a nonequilibrium nuclear configuration because the electronic

FIGURE 28.2 Jablonskii diagram showing the different radiative and nonradiative transitions in a molecule uponexcitation into a singlet state, Sn: S0 = singlet ground state; S1 = first excited singlet state; T1 = first excited tripletstate; Tm = excited triplet state; ISC, ISC1, and ISC2 = intersystem crossings; A = absorption; F = fluorescence; IC =internal conversion; P = phosphorescence; and VR = vibrational relaxation.

VR

VR

PFA VRIC

IC

VR

S1

Sn

S0

T1

TmISC1

ISC2 VR

©2003 CRC Press LLC

Page 6: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

absorption process is so rapid that the atomic nuclei do not have time to change positions or moments.Thus, the adiabatic (Born-Oppenheimer) approximation is often applied to solve the Schrödinger equa-tion (Equation 28.1) and partially separates the eigenstates of the electronic (“fast”) and nuclear (“slow”)subsystems. The intensity of the absorption (i.e., fraction of ground-state molecules promoted to theelectronic excited state) depends on the intensity of the excitation radiation (i.e., number of photons)and the probability of the transition with photons of the particular energy used. A term often used tocharacterize the intensity of an absorption (or induced emission) band is the oscillator strength, f, whichmay be defined from the integrated absorption spectrum by the relationship

, (28.2)

where εν is the molar extinction coefficient at the frequency ν.Oscillator strengths of unity or near unity correspond to strongly allowed transitions, whereas smaller

values of f indicate smaller transition dipole matrix elements of forbidden transitions.

Vibrational Relaxation (VR)In condensed media, the molecules in the Sn state deactivate rapidly, within 10–13 to 10–11 s via VRprocesses, ensuring that they are in the lowest vibrational levels of Sn possible as described by the thermalBoltzmann distribution. Because the VR process is faster than electronic transitions, any excess vibrationalenergy is rapidly lost as the molecules are deactivated to lower vibronic levels of the corresponding excitedelectronic state. This excess VR energy is released as thermal energy to the surrounding medium.

Only under special circumstances — for example, in the gas phase at very low pressures (<10–6 torr)— may emission be observed from higher vibrational levels of the excited state. These emission mech-anisms, known as single-vibronic-level (SVL) luminescence processes, do not generally occur in con-densed media such as tissues.

Internal Conversion (IC)From the Sn state, the molecule deactivates rapidly to the isoenergetic vibrational level of a lower electronicstate such as Sn–1 via an IC process. IC processes are transitions between states of the same multiplicity.The molecule subsequently deactivates to the lowest vibronic levels of Sn–1 via a VR process. By a successionof IC processes immediately followed by VR processes, the molecule deactivates rapidly to S1. As a result,a molecule may be excited to S1 or higher excited states, Sn, depending on the excitation energy used,but the emission takes place only from the lowest excited electronic state, S1. This is known as Kasha’srule. Fluorescence emission from upper electronic states (above S1 or T1) of polyatomic molecules,however, is a rare phenomenon in the condensed phase.

After vibrational relaxation or internal conversion, the thermal population at a given electronic stateis determined by the Boltzmann distribution, which gives the relative population of species in a givenelectronic or vibronic state as compared with those in all other states. The Boltzmann distribution isexpressed by

(28.3)

where ni is the number of molecules in electronic or vibronic state I, gi is the statistical weight of stateof that state, Ei is the energy of state I, k is the Boltzmann constant, and T is the temperature.

Fluorescence (F)Relaxation of the molecule from the S1 state may occur by one of a several different of processes, including:(1) further deactivation to S0 in a radiationless fashion via internal conversion (IC) and VR processes or(2) emission of a photon without a change in spin multiplicity. The latter S1 → S0 transition is known

f d= × − ∫4 315 10 9. ε νν

n

n

g E kT

g E kTi

j

j

i i

j i

j

∑ ∑=

−( )−( )

exp

exp

©2003 CRC Press LLC

Page 7: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

as fluorescence. The energy of the resulting photon corresponds to the difference in energy between thelowest vibrational level of the excited state and the vibrational level of the ground state. Thus, thefluorescence spectrum primarily provides information about the vibrational structure of the ground state.

These radiative transitions are electric-dipole in nature. The probability of a radiative transition(Si → Sj) is proportional to the square of the matrix element, |(M)ij|2, where M is the electric dipolemoment defined by

(28.4)

where e is the electronic charge, rq is the position vector of electron q, and ψ (Si) is the wave function ofelectronic state Si. For fluorescence (i = 0, j = 1) the electronic transition corresponds to M1,0

2.

Intersystem Crossing (ISC)Another process through which molecules in the Si state may relax is through a transition to somevibrational level of the triplet manifold via a mechanism known as an intersystem crossing (ISC) process(ISC1 transition shown in Figure 28.2). The ISC process involves a change in spin multiplicity. Themolecule then relaxes to the lowest vibrational level of T1 via successive IC and VR processes.

From this T1 state, the molecule may return to the ground state S0 either by a radiationless deactivationpath (ISC2 process shown in Figure 28.2) or by the emission of a photon. This latter radiative transitionis known as phosphorescence.

The radiationless deactivation that quite often competes with phosphorescence is the reverse ISCprocess (ISC2). This process, which is often dominant, determines the observed phosphorescence lifetime.The nonradiative rate constant corresponding to the ISC process T1 → S0 is strongly dependent on themagnitude of the energy gap ∆E(T1 → S0). The rate constant decreases exponentially with increasing ∆E;this feature is known as the energy gap law. The energy gap law not only is specific to ISC but also appliesto other nonradiative transitions such as IC.

Phosphorescence and the Triplet StateThe triplet state manifold consists of states T1, T2,… Tn, which have energy levels lower than the corre-sponding singlet states S1, S2,… Sn. This is a consequence of Hund’s rule, which states that the energyassociated with a state having parallel spins is always lower than that of the corresponding one withantiparallel spins. Qualitatively, Hund’s rule may be explained by the tendency for electrons with parallelspins to be further separated in space (i.e., to lie in different molecular orbitals) and consequentlyexperience less coulombic interaction. As a result of Hund’s rule, phosphorescence occurs at longerwavelengths than does fluorescence.1,2

Properties of singlet and triplet states are significantly different. The singlet state is diamagnetic, whereasthe triplet state is paramagnetic. The nomenclature “singlet” and “triplet” is derived from multiplicityconsiderations of the energy level splitting that occurs when the molecule is exposed to a magnetic field.Under the application of an external magnetic field, the triplet state with a spin magnetic momentumsplits into three Zeeman levels. The singlet state, which has no magnetic momentum, is unchanged.

Spin–Orbit CouplingProduction of the triplet state in some compounds is important to photosensitization in certain biomed-ical applications, such as photodynamic therapy (PDT). A process that leads to the production of thetriplet state is spin–orbit coupling. Transitions between different excited singlet states of the samemultiplicity (Sn → Sm or Tn → Tm) may occur easily, but transitions between pure spin states of differentmultiplicities (Sn → Tn) are forbidden by quantum mechanics under the spin-selection rule (∆S = 0).Spin-forbidden transitions, however, do occur under certain conditions. If the spin and orbital motionsof the electrons are independent of one another, triplet–singlet transitions do not give rise to electric

M S S S e Si j i q

q

j→( ) = ( ) ( )∑ψ ψr

©2003 CRC Press LLC

Page 8: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

dipole radiation because of the spin selection rule. However, the spin and orbital motions are actuallynot independent. Basically, the occurrence of spin-forbidden transitions — for example, singlet–triplettransitions — is made possible by coupling of the electron spin with the orbital angular momentum, anonclassical phenomenon that produces a quantum mechanical mixing of states of different multiplicities.Qualitatively, this mechanism may be explained by the fact that the orbital motion of the electron inducesa magnetic field that interacts with its spin magnetic moment. This interaction results in a change of thedirection of the spin angular momentum of the electron.2 This phenomenon is known as spin–orbit(S–O) coupling.

The S–O coupling mechanism actually mixes some singlet character into triplet states and vice versa.This process therefore removes the spin-forbidden nature of the transitions between pure-spin states.Because of the importance of S–O coupling in understanding the phosphorescence process, it is essentialto consider briefly the theoretical treatment of S–O coupling.

General Considerations for Nonradiative PropertiesThe VR, IC, and ISC processes are known as “radiationless transitions.” In the framework of the Born-Oppenheimer approximation, in which the nuclear and electronic motions are assumed to be indepen-dent of each other, the probability of Wmn of a radiationless transition between two electronic states nand m can be treated by perturbation theory and is given by the following expression:1,2

(28.5)

where h is Planck’s constant, θmn is the vibrational wave function i of the electronic state m, θm is theelectronic wave function of the electronic state m, τ is the relaxation time of the vibronic levels, H′ isthe perturbing Hamiltonian and Πij is the product over all vibronic states i and j.

For radiationless transitions between states of similar multiplicity — for example, VR and IC processes— H′ may be the electron–electron repulsion term or the vibronic interaction term. For radiationlesstransitions between states of different multiplicity — for example, ISC processes — H′ is the spin–orbitcoupling term.

A common expression that describes the probability Wmn is the so-called Fermi Golden Rule for thestatistical limit of a dense manifold for the final states. In this case Wmn is given by

where Vmn is the matrix element of the perturbation between the initial state m and final state n, ρ is thedensity of the final state, and h is Planck’s constant.

Delayed Fluorescence (DF)Fluorescence and phosphorescence are the two most common luminescence processes. A less commonemission process is delayed fluorescence (DF). With some chemical systems, the molecule in the tripletstate T1 reverts back to the excited singlet state manifold. Because T1 is always of lower energy than S1 ,

this transition requires some additional activation energy. Delayed fluorescence may occur from the S1

state, exhibiting a spectrum identical to conventional fluorescence (often called “prompt fluorescence”)but having a longer lifetime due to the additional stay in the triplet state. Two types of DF processes canbe differentiated: (1) an eosin-type (E-type) DF produced by repopulation of S1 from the triplet state bythermal activation, and (2) a pyrene-type (P-type) DF produced when pairs of triplet state moleculesinteract, providing an activation energy greater than or equal to the S1 energy. At room temperature,E-type DF can be an important mechanism and has been observed for many organic compounds adsorbedon solid substrates.

Wh

Hmn mi nj m n

ij

= ′∏8 2

2

2 2π τ θ θ Φ Φ

Wh

Vmn mn= ⋅2π ρ

©2003 CRC Press LLC

Page 9: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

28.3 Characterization of Luminescence

Several physical observables may be used to characterize fluorescence and phosphorescence. These includethe following:

1. Emission, excitation, and synchronous spectra2. Quantum yields3. Lifetimes4. Polarization

28.3.1 Emission, Excitation, and Synchronous Spectra

It is first important to determine the positions and nature of the energy levels of the electronic states involvedin the luminescence excitation and emission processes because these energy levels are characteristic propertiesof the compounds of interest. In general, the simplest method of studying the energy levels of the excitedstate is absorption spectroscopy. An alternative method for investigating the absorption process forluminescent compounds is excitation spectroscopy.

The general relationship between the absorption and emission spectra and the vibrational levels ofthe electronic states is illustrated in Figure 28.3. Because a fluorescence emission spectrum is due toradiative decay from Si to different levels of S0, the emission spectrum exhibits the vibrational frequenciesthat correspond to the vibrations of the molecule in its ground state, whereas the absorption spectrumexhibits frequencies of the excited state. The intensities of the vibrational bands are determined inquantum theory by the magnitude of the wave-function overlap (Franck–Condon coefficients) for thevarious vibrational levels in the ground and excited states (Figures 28.3B and 28.3C). The overlapcoefficients are the Franck–Condon factors.

Figure 28.3A shows the potential energy curves of the excited and ground states and the transitionscorresponding to an excitation (or absorption) and emission process. Figures 28.3B and 28.3C show twotypical cases of electronic states with their potential minima at different relative positions on the inter-nuclear distance coordinate R. In Figure 28.3B, the strongest transition is the 0–0 transition, at whichthe overlap is the largest. In Figure 28.3C, the most probable transition is the 0–4 transition involvingthe vibrational level ν′ = 4 of the ground state and the vibrational level ν′ = 0 of the excited state. Theenvelopes of the vibrational progressions illustrated in Figures 28.3B and 28.3C depend on the relativedisplacement δR between the potential minima of the excited and ground states; for the simple case inwhich the ground vibrational level is thermally populated, the shape of the envelopes is determined bythe Pekarian formula. For δR = 0, there is no vibrational progression; for large δR, the Pekarian distri-bution becomes a Gaussian distribution. Examination of the vibronic structure of the fluorescence, orphosphorescence, spectrum can therefore yield important information about the potential curves of thesinglet and ground states or triplet and ground states, respectively.

With conventional emission (excitation) spectra, the excitation (emission) wavelength is fixed whilethe emission wavelength is scanned over the spectral region of interest. With synchronous spectra,excitation and emission wavelengths are scanned synchronously while maintaining a constant wavelengthinterval between them.3,4

28.3.2 Quantum Yields

Emission quantum efficiencies, also known as quantum yields, determine the sensitivity of the lumines-cence measurements. Quantum efficiencies are intrinsic molecular parameters of a compound undergiven conditions of temperature, solvent, and other environmental factors.

The fluorescence quantum efficiency ΦF is defined as:

(28.6)ΦF = Number of luminescence photons

Number of photons absorbed

©2003 CRC Press LLC

Page 10: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

The phosphorescence quantum efficiency ΦP is defined in a similar manner. Quantum efficiencies canalso be defined for processes other than emission and are used to define the fraction of molecules thatundergoes a specific process.

28.3.3 Lifetimes

Another means for characterizing a luminescence emission process is the determination of its lifetime.The luminescence lifetime, τ, is defined as the time required for the emission to decrease to 1/e of itsoriginal intensity following a δ-pulse excitation.

If I(t) is the luminescence intensity at time t and I0 is the intensity at t = 0, then τ is given by:

IL(t) = I0 exp(–t/τ) (28.7)

FIGURE 28.3 (A) Diagrams of the potential energy curves of two electronic states showing the absorption andemission transitions between these two states and the typical intensity distribution of the corresponding spectra.(B) Potential energy curves with unchanged potential minima. The most intense transition is 0–0 (maximum overlapof the vibronic wave functions). (C) Potential energy curves with potential minima shifted with respect to each other.In this example, the most intense transition is 0–4.

E

A

I

I I

E E

B C

R

ν = 4ν = 3

ν = 2ν = 1

ν = 0

ν′= 4

ν = 0

E(ν~)

E(ν~)E(ν ~)

ν ′= 0

ν = 0

ν ′= 4

ν = 3ν = 2

ν = 1ν = 0

ABSORPTIONEMISSION

RR

0 4 62 0 4 6 8 102

©2003 CRC Press LLC

Page 11: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

It is important to differentiate the intrinsic natural lifetime, τ*, from the observed lifetime, τ, for agiven radiative process. The intrinsic natural lifetime, τ*, is measured only when there is no radiationlessdeactivation process competing with the emission — that is, when ΦL = 1 for the resulting luminescence.

Experimentally, fluorescence can be differentiated from phosphorescence by its shorter lifetime. Flu-orescence lifetimes of organic molecules are on the order of 10–9 to 10–7 s, while phosphorescence lifetimesrange from 10–3 s to several seconds. Table 28.1 compares the oscillator strengths and radiative lifetimesof fluorescence (S1 → S0) and phosphorescence (T1 → S0) emission and absorption of different origins.

28.3.4 Polarization

Polarization is another physically observable characteristic of luminescence. Polarization is caused byunique symmetries and orientations of electric moment vectors and wavefunctions involved in electronictransistions. The electric dipole moment determines the direction along which charge is displaced in amolecule undergoing an electronic transition. It is possible to study the polarization of the transitionusing polarized light to excite and detect luminescence.

A common method for determining the degree of polarization, P, is photoselection. In this method,one excites the sample with polarized light and measures the luminescence intensity along two perpen-dicular directions. The degree of polarization is defined as

(28.8)

where IEE and IEB are the luminescence intensities measured along the direction parallel and perpendicularto the excitation electric vector, E, respectively. P is related to the angle, θ, between the absorption vectorand the emission vector by the relationship

(28.9)

If the absorption and emission vectors are parallel (θ = 0°), P = 0.5. If these vectors are perpendicular(θ = 90°), P = –0.33. In practice, P has a value between these two limiting cases.

28.4 Biomedical Applications

Biomedical fluorescence spectroscopy is an extremely large and growing field of research. In an attemptto classify the various types of research and clinical studies that are being performed, we have groupedthis field into three categories:

TABLE 28.1 Radiative Lifetimes and Oscillator Strengths in the Electronic Transitions for Polynuclear Aromatic Compounds

Origin of TransitionsRadiative Lifetime

(s) Oscillator Strength

S1 →→→→ S0 Transitions

ππ* 10–7–10–9 10–3–1πσ or σπ* > 10–6 < 10–2

nπ* ∼ 10–6 ∼ 10–6

T1 →→→→ T0 Transitions

nπ* (or σπ* or πσ*) 10–2–10–4 10–7–10–5

ππ* in halo-aromatic 1–10–3 10–9–10–6

ππ* in unsaturated hydrocarbons ∼ 10–2 10–11

PI I

I IEE EB

EE EB

=−+

P =+

3

3

2

2

cos

cos

θθ

©2003 CRC Press LLC

Page 12: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

1. Biochemical analyses of individual compounds2. In vitro analyses3. In vivo analyses

Biochemical analyses are the basis for the fundamental or general studies of individual biochemicalcompounds that form the basic constituents of biological samples (i.e., tryptophan, NAD, NADH,hemoglobin, etc.). From these studies the basic fluorescence properties of common fluorophores aregenerally determined and characterized. The second category, in vitro analyses, is further divided intotwo subcategories: cellular measurement and tissue measurement. These analyses typically provide addi-tional information associated with complex systems such as cells and tissues. The third classificationcategory, in vivo analyses, can also be subdivided into two more subcategories: animal studies and humanclinical studies. These experiments are generally performed only for fluorescence analysis techniquesmeant to be used for optical diagnostic procedures. In addition to classification based upon the type ofanalysis performed, it is also possible to subcategorize based upon the type of analyte measured andwhether it is an endogenous species or an exogenous fluorophore (e.g., used as a label or marker for thetissue area of interest).

28.4.1 Biochemical Analysis of Individual Species

Fluorescence analyses of a wide variety of biochemical species that have been applied to and extractedfrom biological samples have been performed for quite a long time. These analyses are generally classifiedbased upon whether or not the fluorescent species is an endogenous (naturally occurring) or exogenous(externally administered) fluorophore.

28.4.1.1 Endogenous Fluorophores

A large number of endogenous fluorophores, autofluorescent species, exist within biological samples.5–7

The majority of these species are typically associated with the structural matrix of tissues (e.g., collagen8

and elastin9) or with various cellular metabolic pathways (e.g., NAD and NADH).10 In the case ofstructural matrices, the most common fluorophores are collagen and elastin. The resulting fluorescencefrom these compounds is due primarily to the cross linking of various amino acids in their structure,which typically provides the conjugated systems that are generally found in most fluorescent molecules.In addition, because of the various degrees of cross linking that can occur in these species, the resultingfluorescence emission spectrum is typically very broad and featureless, ranging anywhere from approx-imately 325 to 600 nm.

Some of the most intense endogenous fluorophores that exist in humans and animals are involved incellular metabolism. The predominant species in this category include the reduced form of nicotinamideadenine dinucleotide (NADH),11 the various flavins (FAD, etc.),12 and the strongly fluorescent lipopig-ments (e.g., lipofuscin, and ceroids).13,14 In addition to these species, many other endogenous fluorophoresexist, exhibiting emission of various strengths and covering various spectral ranges in the ultraviolet(UV) and visible regions of the electromagnetic spectrum. These fluorophores include aromatics, aminoacids such as tryptophan, tyrosine, and phenylalanine,6 various porphyrins (hemoglobin, myoglobin,etc.),6 and in certain cases and locations, red porphyrin fluorescence due to bacteria.10 Figure 28.4 showsthe absorption and fluorescence spectra of various tissue fluorophores.

These biochemical species are generally structural or metabolic, so they can provide a significantamount of information about differences in tissues. One of the most common reasons for investigatingthese fluorescent species is the possibility for diagnosis of various diseases (e.g., cancer) without requiringexogenous fluorescent markers or tags. Because cells in various disease states often undergo different ratesof metabolism, or have different structures, there are often distinct differences in their fluorescent emissionspectra. These differences in fluorescence emission generally depend on at least one of the followingparameters: fluorophore concentration or spatial distribution throughout the tissue, local microenviron-ment surrounding the fluorophores, the particular tissue architecture being interrogated, and a wave-length-dependent light attenuation due to differences in the amount of nonfluorescing chromophores.

©2003 CRC Press LLC

Page 13: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

The first of these reasons, change in fluorophore concentration, can be attributed to many differentevents. In certain cases, various chemicals may not be produced in a diseased tissue. In other cases, thedistribution or form of the fluorophore may vary throughout the tissue. For instance, NADH is anextremely fluorescent molecule in its reduced form but is nonfluorescent in the oxidized form. Therefore,depending on the specific phase of a particular metabolic reaction that may be occurring, the relativefaction of NADH in the reduced and oxidized forms may vary greatly. Another reason for differences inthe fluorescence emission from various tissue types is the difference in the local environment surroundingthe fluorophore.

Change in the microstructure (local environment) surrounding a particular fluorophore can havesignificant effects on its fluorescence properties. Fluorescence properties that are often affected by suchchanges include the fluorescence quantum yield, the spectral position of its fluorescence emission max-imum, its spectral line width, and its fluorescence lifetime. All these factors can have a significant impact

FIGURE 28.4 Absorption (A) and fluorescence emission (B) spectrum of many of the dominant fluorophorespresent in tissue. (Adapted from Wagnières, G.A., Star, W.M., and Wilson, B.C., Photochem. Photobiol., 68, 603, 1998.)

©2003 CRC Press LLC

Page 14: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

on the overall fluorescence emission of the entire tissue sample. In addition to the local environmentsurrounding the fluorophore, the larger macrotissue structure or architecture also plays a significant rolein the resulting fluorescence because of its specific optical properties (e.g., refractive index, scatteringcross sections). This feature is described in more detail in Section I (Photons and Tissue Optics) chaptersof this handbook.

An important factor that often plays a significant role in the differences associated with various diseasestates of tissue is the presence of nonfluorescing chromophores (hemoglobin, etc.). These chromophores,which can vary in concentration depending upon the state of the tissue, can absorb various wavelengthsof fluorescent light emitted from the tissue or even absorb specific wavelengths of the excitation lightused for the analysis. One example of this case is in the fluorescence diagnosis of tumor tissue, whichtypically has an increase in vascularization over normal tissues due to angiogenesis. Because of thisincrease in vascularization, the increased presence of hemoglobin often causes increased absorption oflight in the visible region of the electromagnetic spectrum.

Due to this complex nature of biological tissues, extensive studies have been performed on individualspecies at the fundamental biochemical level. These fundamental studies of the absorption and fluores-cence properties of various biochemical fluorophores give us a better understanding of the complexinteractions that may be occurring in tissue samples.

In addition to conventional spectrally resolved fluorescence analyses, which are capable of providinga significant amount of information about the various fluorophores present, as well as some informationabout their local environment, some work in the field of time-resolved fluorescence analyses has alsotaken place. An in-depth analysis of this subject can be found in Chapter 9 of this handbook. Time-resolved spectroscopic analyses can provide information about molecular interactions and motions thatoccur in the picosecond–nanosecond time-scale range. Understanding these interactions can be especiallyuseful in the analysis of biomolecular structure and dynamics. Recent advances in time-resolved fluores-cence spectroscopy in the field of biological analyses have led to a better understanding of the origin ofnonexponential fluorescence decay in proteins, the use of tryptophan analogs as unique spectroscopicprobes of protein–protein interactions, the detailed characterization of protein-folding processes andintermediates, the development of new approaches to the study of DNA-protein interactions,15 and theanalysis of subdomains in proteins.16,17

28.4.1.2 Exogenous Fluorophores and Molecular Markers

In addition to the large number of endogenous fluorophores that have been used for biological diagnostics,many different exogenous fluorophores have been created and studied as well. These fluorophores havebeen created for many different purposes and applications in the field of biological monitoring, rangingfrom monitoring cellular function using fluorescent reporter dyes or molecules for various biochemicalspecies (Ca+2, Mg+2, pH, nucleic acid sequences, etc.) to the demarcation of tumors with a relatively recentclass of exogenous fluorophores. In the case of the monitoring of cellular function and chemical distri-bution, a large number of dyes with varying fluorescence properties for most of the more common cellularspecies can be obtained commercially from companies such as Molecular Probes, Inc. (Eugene, OR).These fluorescent reporter dyes are typically used to monitor the distribution of important chemicalspecies throughout a cell by obtaining fluorescence microscopy images after injecting it with the dye. Inaddition to simply monitoring the distribution of certain chemicals, several dyes can be used to determinethe viability of the cells or the permeability of their membranes. A more detailed description of thesedyes and the types of analyses of which they are capable is presented in Section 28.4.2.

A second type of fluorescent reporter dye developed recently is known as a molecular beacon.18–21

These molecular beacons are constructed by attaching a fluorescent dye (e.g., fluorescein) to one end ofan oligonucleotide sequence, and a quencher molecule (e.g., DABSYL) to the other end. The particularoligonucleotide sequence chosen for the molecular beacon comprises the complementary sequence tothe RNA or DNA sequence to be measured. For the molecular beacon to function properly, the lastseveral nucleic acids on either end of the oligonucleotide strand must be complementary to each other.This forces the molecule to assume a hairpin form in its native state that brings the quencher molecule

©2003 CRC Press LLC

Page 15: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

in close proximity to the fluorophore of the other end. The proximity of the quencher molecule to thefluorophore prevents the fluorophore from emitting any fluorescent light. However, when the molecularbeacon is in the presence of its complementary sequence to the remainder of the oligonucleotide, thebeacon is opened up, thus separating the quencher and fluorophore molecules and allowing the fluores-cent dye to emit. This feature is described in more detail in Chapter 57 of this handbook.

In addition to fluorescent reporter dyes and molecular beacons, a third major area of research inbiologically useful exogenous fluorophores has been in the area of tumor demarcation or sensitization.The most common of these tumor markers also act as sensitizers, or tumor-killing agents, for a uniquescheme of cancer treatment known as photodynamic therapy (PDT). (A much more detailed descriptionof PDT is offered in Chapter 36 of this handbook.) Some of the most commonly used and best charac-terized of these photosenstizers are hematoporphyrin derivatives (HpD),22,23 phenophorbide-a,24

mTHPC,25 benzoporphyrin derivatives (BPD), 26,27 tin etiopurpurin (SnET2),28 hypericin,29,30 and 5-ami-nolevulinic acid (ALA).31–35 HpDs (i.e., Photofrin) are the best studied of these compounds.

HpDs were derived from the functionalization of hematophorphyrins (Hp) with hydrophobic substi-tutents that would allow these compounds to be taken up more readily in the tumor tissues than theunsubstituted Hps.36 After these compounds had been developed, it was found that they could also serveas photosensitizers because they are capable of being excited and activated by the absorption of visiblelight. Although HpDs are still used quite extensively today for PDT treatments, many other compoundsor classes of compounds with more appropriate characteristics for clinical drugs (such as those listedpreviously) have been developed.

The most desirable optical properties of PDT sensitizers are strong absorbtivity in the red or near-infrared (NIR) region of the electromagnetic spectrum and a high triplet-state quantum yield. The firstof these two properties is important for several reasons. One reason is that the use of red or NIRwavelengths of light for excitation of the sensitizer causes much less damage to nearby healthy tissue. Atred and NIR wavelengths, naturally occurring chromophores in the tissue absorb little light. In addition,because the absorption of these wavelengths by natural chromophores is minimal, the resulting back-ground fluorescence from nearby healthy tissue is minimized, providing a much more accurate tumormarker as well as sensitizer. A third potential benefit gained by having photosensitizers with strongabsorption spectra in the red and NIR is the ability to destroy tumors under thin layers of normal tissue.

When photosensitizers activated with visible or ultraviolet light are used, the absorption cross sectionof the healthy overlaying tissue is too great to allow a significant fraction of the light to penetrate deeperthan approximately 100 µm. However, lower-energy photons, such as those in the red or NIR, penetratesignificantly and could allow for the activation of sensitizers in tumors below healthy overlaying tissue.In addition to the desire to develop new sensitizers and markers with red-shifted absorption profiles, itis also important to obtain a high triplet-state conversion rate in these molecules. Triplet-state conversionis very important to formation of the reactive oxygen species responsible for the drug’s activity.10

28.4.2 In Vitro Analyses and Diagnostics

In vitro analyses can typically be classified in two different categories: cellular analyses and tissue analyses.In vitro analyses are important for gaining a greater understanding of basic fundamental biologicalprocesses that are occurring and understanding any potential interferences or problems that may bepresent when investigating more complex biological systems (i.e., living animals).

In addition to gaining better fundamental biochemical understanding, in vitro fluorescence analysesare often performed to better understand the interactions or interferences that often take place in complexsystems as opposed to individual chemicals. By investigating these effects on in vitro samples such as cellsor tissues, investigators can determine potential diagnostic procedures that could be performed in vivoor identify problems with potential diagnostic procedures prior to involving live subjects. When per-forming analyses on neat biochemical systems, it is not possible to investigate the interactions of variousbiochemicals with others that are present in functional systems (i.e., cells, tissues, etc.). In addition, the

©2003 CRC Press LLC

Page 16: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

effect of the local environment on the optical properties of the various fluorophores can be investigatedmuch more accurately by using in vitro systems as compared to biochemical analyses alone.

28.4.2.1 Cellular Analyses

Cells represent one of the most basic of biological systems capable of performing chemical reactions.Monitoring endogenous as well as exogenous fluorophores on the cellular level has provided a great dealof information about many different processes. Several different fluorescence techniques have beendeveloped for the performance of such analyses. These techniques include probing with chemical orbiochemical sensors as well as obtaining fluorescence images using any of several different forms offluorescence microscopy. In this section, we provide a brief review of the various types of cellular processesthat have been investigated using fluorescence spectroscopy.

Several other chapters in this handbook provide a more detailed description of the techniques andtheir applications. Chapter 54 discusses living cell analysis using optical techniques and provides a generalreview of various optical methods used for analyzing living cells. Chapter 20 on biosensors in medicalapplications, Chapter 59 on PEEBLE nanosensors for in vitro bioanalysis, and Chapter 60 on nanosensorsfor single-cell analysis provide detailed descriptions of chemical and biochemical sensors used for cellularanalyses. Chapter 10 on confocal microscopy, Chapter 11 on two-photon excitation fluorescence micros-copy, and Chapter 9 on lifetime-based imaging provide detailed reviews of the theory and applicationof fluorescence microscopy.

28.4.2.1.1 Autofluorescence of CellsThe vast majority of fluorescence microscopy is performed with the use of exogenous reporter dyes;however, a growing field of fluorescence microscopy relies on autofluorescent species inside the cells fordiagnostic information. Such autofluorescence measurements provide an important tool for biomedicaldiagnostics. Much of the research in autofluorescence microscopy is performed as preliminary researchprior to testing in vivo diagnostic procedures or as a medical diagnostic procedure itself. Some recentexamples are described next.

Cell Proliferation and Cell Analysis — One of the most sought-after applications of fluorescence spectros-copy in biomedical optics is the ability to distinguish normal tissue from cancerous or even precanceroustissue in a real-time clinical setting, without administering any drugs. In order to determine the potentialof this technique, several researchers have sought to answer the questions of whether or not uniquefluorescence spectral patterns were associated with cell proliferation and whether differences betweenrapidly growing and slowly growing cells could be identified. Native cellular fluorescence was used toidentify terminal squamous differentiation of normal oral epithelial cells in culture (Figure 28.5).37

In one such analysis, fluorescence excitation spectra were capable of distinguishing between slow- andrapidly growing cells in three different types of cells. To perform the analyses, excitation spectra wereobtained by scanning the spectral range of 240 to 430 nm while monitoring the fluorescence emissionat 450 nm. By taking the ratio of the intensity of the major broad-band peak at 320 to 350 nm to a pointon the down slope of the curve at 370 nm, investigators found a statistical difference between slow- andrapidly growing cells. In addition, it was also possible to distinguish the slow-growing cells from therapidly growing cells by obtaining fluorescence emission spectra of the various cells with an excitationwavelength at 340 nm and an emission wavelength range of 360 to 660 nm. These results demonstratea great potential for discrimination of proliferating and nonproliferating cell populations in vivo.38

In a similar study, laser-induced fluorescence, with excitation by the 488-nm laser emission line of anargon ion laser, was used to differentiate between normal and tumor human urothelial cells. Experimentswere performed using a confocal microspectrofluorimeter, allowing individual cells to be probed; thebroadband autofluorescence emission between 550 and 560 nm, corresponding to oxidized flavoproteins,was monitored. An analysis of the data showed that the maximum autofluorescence intensity of normalurothelial cells was approximately ten times higher than that of any of the tumor cell types tested, therebysuggesting that the concentration of an oxidized flavoprotein in tumor urothelial cells is significantly less

©2003 CRC Press LLC

Page 17: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

than in a normal urothelial cell. Not only could this observation lead to the development of a clinicaldiagnostic procedure for the real-time differentiation between normal, cancerous, and potentially pre-cancerous urothelial tissues, but it also provides a better understanding of the biochemical differencesbetween normal and cancerous tissues.39

Synchronous luminescence (SL) is a unique approach that could improve the selectivity of fluorescencetechniques for cancer diagnostics. As discussed previously, autofluorescence of neoplastic and normaltissues has been observed using fixed-wavelength laser excitation. However, the use of fixed excitationmight not be sufficiently selective in some diagnostic applications due to strong overlap of the emissionspectra from different fluorophores. An alternative approach is the SL method, which involves scanning

FIGURE 28.5 Native cellular fluorescence of normal oral epithelial cells in culture. This figure shows several nor-malized fluorescence excitation spectra of normal oral epithelial cells that have been grown in several different media:KGM (dashed line), DMEM/F12/FCS (solid line), and DMEM/F12/NaCl (dotted line). The various culturing mediawere used to induce different stages of squamous differentiation in the cells, with KGM providing a medium fornormal growth without differentiation, DMEM/F12/FCS providing a medium known to cause cell stratificationwithin keratinocytes, and DMEM/F12/NaCl providing a medium capable of inducing the terminal stage of squamouscell differentiation in keratinocytes. The fluorescence excitation scans were taken under the following conditions:(A) λex 200 to 360 nm; λem 380 nm; (B) λex 240 to 415 nm; λem 450 nm; (C) λex 250 to 420 nm; λem 480 nm; (D) λex

270 to 480 nm; λem 520 nm. From such spectra, it is possible to differentiate between the various states of the cellsin vitro. (Adapted from Sacks, P.G. et al., Cancer Lett., 104, 171, 1996.)

Nor

mal

ized

Inte

nsity

Nor

mal

ized

Inte

nsity

Nor

mal

ized

Inte

nsity

Nor

mal

ized

Inte

nsity

1.0

0.8

0.6

0.4

0.2

0.0

1.0

0.8

0.6

0.4

0.2

0.0

1.0

0.8

0.6

0.4

0.2

0.0

1.0

0.8

0.6

0.4

0.2

0.0

Wavelength (nm)Wavelength (nm)

Wavelength (nm)Wavelength (nm)

200 220 240 260 280 300 320 250 270 290 310 330 350 370 390 410 430

240 260 280 300 320 340 360 380 400 420 270 300 340 380 420 460 480

A C

DB

©2003 CRC Press LLC

Page 18: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

excitation and emission wavelengths simultaneously while keeping a constant wavelength interval betweenthem.40,41 This method, also referred to as synchronous fluorescence (SF), has been developed for mul-ticomponent analysis and used to obtain fingerprints of real-life samples, as well as for enhancingselectivity in the assay of complex systems. This SF procedure has been shown to simplify the emissionspectrum and provides for greater selectivity when measuring fluorescence or phosphorescence frommixtures of compounds.3,4,42,43

Spectral differences in SF emission profiles are related to the specific macromolecules that differedbetween neoplastic and normal cells. The SF technique has been shown to improve spectral selectivityin detecting normal and cancer cells for potential use in biomedical diagnostics.43 Whereas conventionalfixed-excitation fluorescence could not show any differentiation between normal and cancerous cell lines,spectral difference between the fluorescence spectra of the normal rat liver epithelial (RLE) and neoplasticrat hepatoma McA cell lines were detected using SF. The results demonstrated the great potential of SFas an improved screening tool for cancer diagnosis in specific cases where conventional fixed-excitationmethods are not sufficiently effective.

Biological Fluids, Semen, Seminal Plasma, and Spermatozoa — In addition to being used for the study ofcancer, autofluorescence microscopy is also being investigated for its potential in many other forms ofbiomedical diagnostics. One potential application involves the determination of sperm viability andmotility for real-time fertility diagnoses. In one study, fluorescence emission spectra of human semen,seminal plasma, and spermatozoa, excited at 488 nm, were obtained over the range of 500 to 700 nm.

Under these conditions, emission peaks from each component were observed at 622 nm. The intensityof the emission peaks from spermatozoa at 622 nm was strongly correlated with the concentration ofspermatozoa (r = 0.837; p = 0.0001). In addition, sperm motility could also be correlated significantly,although to a poorer extent, with the intensity of the fluorescence emission peaks from spermatozoa(r = 0.369; p = 0.019) and semen plasma (r = 0.356; p = 0.024).44 The SF method has been investigatedas a rapid screening tool for monitoring DNA damage (DNA adduct metabolites) and in monitoringbiological fluids in animal studies, thus providing a technique for early cancer prescreening at the DNAlevel.42

28.4.1.1.2 Cellular Fluorescence Using Exogenous DyesFluorescence analyses of living cells by means of exogenous reporter molecules represent the vast majorityof spectroscopic analyses performed on cells. Such analyses can generally be classified into one of twocategories: optical sensor-based analyses or fluorescence imaging-based analyses. Both techniques haveadvantages and disadvantages for measurement of individual chemical species inside a single living cell.Optical sensors allow for accurate determination of chemical concentrations at individual points in thecell, while imaging-based analyses provide a means of monitoring the spatial distribution of the analytethroughout the cell.

Several types of optical sensors have been developed for minimally invasive fluorescence monitoringof individual chemical species inside a single cell.45–52 These sensors are based on the same principles andconcepts as those discussed in the chapter on biosensors in medical applications, but their dimensionsare much smaller. Usually, an exogenous fluorescent dye whose properties (i.e., quantum yield, emissionmaximum, etc.) change in the presence of the analyte of interest is attached to the probing end of thesensor. Such sensors have been used to measure many analytes within living cells, thus providing infor-mation about many different biological processes. These analytes include calcium ions, magnesium ions,nitric oxide, glutamate, lactate, glucose, benzo[a]pyrene tetrol, and even oligonucleotides (i.e., DNA,RNA).43–50 Because this relatively new field of sensors is described in greater detail in this handbook inChapter 59 on PEEBLE nanosensors for in vitro analysis and Chapter 60 on nanosensors for single-cellanalysis, this chapter will not go into any further detail on the subject.

Fluorescence microscopy is the most common technique for monitoring the spatial distribution of aparticular analyte at many different locations simultaneously throughout a cell. In such analyses, one orseveral different exogenous fluorescent dyes are introduced into the cell and allowed to disperse. Afterdispersing throughout the cell, they begin to interact with the analyte of interest; this interaction, in turn,

©2003 CRC Press LLC

Page 19: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

causes change in the fluorescence properties (intensity, spectral shift, etc.) of the dye. Therefore, byobtaining a fluorescence image of the cell at a specific wavelength corresponding to the maximum emissionwavelength of the dye, one can determine the relative concentrations of the analyte at specific locations.

The great amount of information derived from such analyses has allowed researchers and companies(e.g., Molecular Probes) to develop many different fluorescent dyes for a wide variety of compounds.These dyes are available with many different excitation and emission maxima for most analytes, allowingthe experimenter to choose the best dye for the particular application. The dyes most commonly usedtoday for fluorescence microscopy have red or NIR excitation profiles, thus allowing the dye to be excitedwithout providing the energy necessary to excite any of the autofluorescent species present inside thecell. In addition, the lower-energy light also causes less damage to the living cells than would visible orUV wavelengths, which are capable of cross linking DNA or proteins.

Cell Proliferation — Cell proliferation analyses are one of many types often performed using exogenous dyes.Learning about the part of the cell cycle that a particular cell or group of cells is in, or about the way in whichit died (necrosis vs. apoptosis, for example) can provide a great deal of biomedically useful information. Forinstance, the determination of apoptotic cell death is of particular importance in the study of chemicalcarcinogenesis as well as in the development of novel PDT drugs. For this reason, many different types offluorescence analyses have been developed to determine the extent of apoptosis in cells or tissues.

One such method employed a commercially available ApopTag kit (Serologicals Corporation, Norcross,GA) and automated fluorescence image analysis to quantify the distribution of apoptosis in formalin-fixed,paraffin-embedded liver tumor sections from rats whose tumors were induced by 2-acetylaminofluorene.In this work, specific treatments of tissue sections were developed for quenching the autofluorescencebackground from the cells of the tissue sample. Once the autofluorescence had been quenched, propidiumiodide was used as a counterstain for the nuclei. Automated statistical evaluation of the percentage ofnuclei stained positively for apoptosis was determined by using dual fluorescence detection and opticalmicroscopy. The quantitative results indicated that the staining index for apoptosis in normal rat livercells was 0.14 ± 0.04%, whereas well- and poorly differentiated tumor cells showed increases of 3.48 ±0.59% and 7.41 ± 0.81%, respectively.53

Cellular Response — Another important application of fluorescence microscopy using exogenous fluo-rophores is in elucidating the role of particular chemicals in cellular biology and determining theassociated kinetic constants for those processes. One chemical and cellular process that has been inves-tigated using fluorescence microscopy and exogenous fluorophores is protein kinase C (PKC) and its rolein signal transduction of many bioactive substances. In this study, phorbol-13-acetate-12-N-methyl-N-4-(N,N′-di(2-hydroxyethyl)amino)-7-nitrobenz-2-oxa-1,3diazole-aminododecanoate [N-C12-Ac(13)],the fluorescent derivative of 12-o-tetradecanoylphorbol-13-acetate (TPA), was synthesized to monitorthe location of phorbol ester binding sites and evaluate its potential use as a probe for PKC in viablecells. The maximum excitation wavelength of N-C12-Ac(13) is close to 488 nm, making possible the useof an argon-ion laser for excitation of the dye molecule. When incubated with 100 nM N-C12-Ac(13),P3HR-1 Burkitt lymphoma cells accumulated the dye rapidly, reaching a maximum fluorescence (20-fold above the autofluorescence background) within 25 min. The subsequent addition of unlabeled TPAsignificantly decreased the fluorescence of N-C12-Ac(13) in the cells, in a dose-dependent manner,indicating specific displacement of the bound fluoroprobe.

Using this same competitive displacement technique, researchers displaced [3H]-phor-bol-12,13dibutyrate ([3H]-PBu2) from rat brain cytosol with N-C12-Ac(13), which was found to exhibitan apparent dissociation constant and biological activity similar to that of TPA. In addition, like TPA,N-C12-Ac(13) also induced the expression of Epstein-Barr viral glycoprotein in P3HR-1 cells and dif-ferentiation of promyelocytic HL60 cells and caused predicted changes in the mitotic cycle of histiocyticDD cells. Microscopic fluorometric images of single cells for such analyses verified that the bound dyeN-C12-Ac(13) showed bright fluorescence in the cytoplasm and dim fluorescence in the nuclear region,a phenomenon consistent with dye binding mainly to cytoplasmic structures or organelles.54

©2003 CRC Press LLC

Page 20: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

Immunocytofluorometric Analyses — In addition to using exogenous fluorescent dyes to monitor theconcentration or distribution of various chemicals or ions within a cell, investigators have found usefulapplications for fluorescence spectroscopy in the identification of specific types of cells in immunocytof-luorometric analyses. In such analyses, fluorescent molecules are attached to a monoclonal or polyclonalantibody that is specific to a surface protein or a class of surface proteins on a particular type of cell. Thecells to be analyzed are then exposed to these fluorescently labeled antibodies. Once the antibodies haveattached to the cells with the appropriate surface proteins, a fluorescence analysis system (e.g., a flowcytometer) can distinguish the cells of interest from the other cells present on the basis of a fluorescentproperty (e.g., spectral band) of the dye molecule attached to the antibody.

In a recent example, a time-resolved laser-induced fluorescence measurement system was used inconjunction with pyrene-derivatized molecules to perform immunocytofluorometric analyses. By usingthese pyrene-derivatized molecules [N-(1-pyrene)maleimide, 1-pyrenesulfonyl chloride and 1-pyre-neisothiocyanate], fluorescent lifetimes for the labeled antimouse-IgG were in the range of 20 to 55 ns.Due to this long lifetime, it was possible to distinguish these cells easily from other nonlabeled cells whosefluorescence lifetimes were much shorter.55

28.4.2.2 Tissue Analyses and Diagnostics

Many studies have been performed on tissue samples7,40,41,56–62 to help us understand the interactionbetween cells and to bring us closer to the much more complex area of in vivo analyses. Tissue studiesprovide a platform much closer to true in vivo analyses in terms of structural architecture on microscopicand macroscopic scales than do cellular analyses. Ex vivo tissues provide an excellent starting point forthe development of many in vivo diagnostic procedures (e.g., optical biopsy). In addition, when comparedwith in vivo systems, ex vivo tissue samples are much easier to handle and manipulate.

Over the last two decades, many different types of fluorescent tissue analyses have been performed ona wide variety of samples. The types of tissues range from mouse and rat tumors41 to human teeth.60

Instrumentation has ranged from commercially available fluorescence microscopes to laboratory-builtresearch instruments for specialized measurements. The development of fiber-optic instrumentation hasmade it possible to perform analyses in any area accessible to an endoscope.

28.4.2.2.1 Autofluorescence of TissuesMany fluorescent species are present in biological tissues, and we want to develop diagnostic techniquesthat do not require chemical pretreatment of tissues. Therefore analyses based on autofluorescencerepresent a large portion of the fluorescence diagnostic procedures being developed. Some of the morepromising autofluorescent diagnostic results as well as their state of development are described in thefollowing sections.

Cancer Diagnostics — Recently, there has been a great deal of interest and research in the developmentof rapid, fluorescence-based clinical oncology.6,10 Many different techniques have been developed for suchanalyses in different types of tissues and have shown a great deal of promise. These techniques and theinstrumentation used for these measurements are described in greater detail in the following sections ofthis chapter. The effectiveness of the techniques is often determined by comparing their results withhistological data, which are considered the “gold standard.” Based upon the results of these comparisons,we can calculate three quantitative values to provide an objective comparison of various techniques.

Calculation of these three quantities (sensitivity, specificity, and accuracy) depends upon classifying aparticular analysis as either a true positive (TP), which represents classification by both techniques asmalignant; a false positive (FP), which corresponds to a classification as malignant by fluorescencespectroscopy and normal by histology; a false negative (FN), which corresponds to a normal classificationby fluorescence and a malignant classification by histology; or a true negative (TN), which correspondsto classification by both techniques as normal. Once the appropriate classifications have been performed,sensitivity, specificity, and accuracy are calculated as follows:

©2003 CRC Press LLC

Page 21: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

Sensitivity = TP/(TP + FN)Specificity = TN/(TN + FP)Accuracy = (TP + TN)/(TP + TN + FP + FN)

An example of a laboratory-constructed instrument for tissue analyses is given by Alfano and co-workers, who developed a system for the differentiation of cancerous and normal human breast tissuesand lung tissues.61,62 In this work they developed a system that uses a pulsed or a continuous-wave (CW)excitation source for the differentiation of normal and cancerous tissues., The fluorescence emissionspectrum for normal tissue caused by excitation with 488-nm light from an argon ion laser was foundto be quite different from that for tumor tissue. The resulting spectra from tumor tissues had smoothemission curves with a maximum at approximately 530 nm. The resulting emission spectra from normaltissues appeared to have three peaks, at 530, 550, and 590 nm. In another study, Feld and co-workersattempted to determine the optimal excitation wavelength for distinguishing between normal and tumortissues based upon their biochemical and histomorphological (architectural) components. They foundthat the most marked differences occurred when an excitation wavelength of 410 nm was used. Usingthis wavelength, they correctly diagnosed 20 of 22 samples studied.63 This optimal wavelength, determinedwith in vitro studies, is consistent with results obtained by Vo-Dinh and co-workers from in vivo clinicalstudies of gastrointestinal cancer diagnostics using laser-induced fluorescence.64,65

Cervical Cancer — In the field of in vitro fluorescence diagnostics for the detection of cervical cancer,there is a relatively small amount of information. However, results that have been published forecast apromising future for this technique. In one such study, Richards-Kortum and co-workers investigatedfluorescence excitation–emission matrices (EEMs) to analyze 18 cervical biopsies from 10 patients.66 Atall excitation–emission maxima, most prominently at 330-nm excitation and 385-nm emission, theaverage normalized fluorescence intensity of histologically normal tissue was significantly greater thanthat of histologically abnormal tissue. A diagnostic algorithm based upon this relative intensity differencewas developed to differentiate between histologically normal and abnormal biopsies with a higher sen-sitivity but a lower positive predictive value and specificity than colposcopy. However, when comparisonsof histologically normal and abnormal biopsies from the same patient were performed, sensitivity resultsof 75%, positive predictive value results of 86%, and specificity results of 88% were achieved for thespectroscopic identification of histological abnormality. These results compare favorably with colposcopyresults. Based on these results, in vivo studies of cervical tissue fluorescence have also been conducted.66

In addition to research demonstrating the ability to distinguish between normal cervical tissues andinvasive carcinomas in cervical tissue, research has also attempted to distinguish between dysplasia andinvasive carcinomas. To this end, a technique developed recently for the in situ detection of melanomashas been applied for determining in vitro dysplasia and invasive carcinomas in the cervix uteri. Cervixuteri exhibit a fluorescence band with a peak at about 475 nm upon excitation with 365 nm light. At afluorescence intensity of 475 nm, the fluorescence intensity increases concomitantly with the degree ofdysplasia, ranging from 30 counts/100 ms (healthy) to approximately 200 counts/100 ms (carcinomain situ 3). At the rim of a malignancy, the intensity is 250 counts/100 ms and higher. The excitation andemission spectra of the tissue suggest that the endogenous chromophore responsible for the observedfluorescence is NADH.67

Colon Cancer — Another major form of cancer that has seen a significant amount of research andadvancement in terms of in vitro fluorescence diagnosis is colon cancer. In one study, unstained frozensections of colon were studied by fluorescence microscopy to determine which structures fluoresce andto what extent in normal colon tissue and colonic adenomas. Tissues were excited by a 351- to 364-nmemission from an argon ion laser. The resulting fluorescence signals from various locations in the tissuesamples were correlated to the tissue morphology at those locations via histological analyses with con-ventional stains (hematoxylin and eosin [H&E]), Movat pentachrome, mucicarmine, and oil red O dye).

In normal colon tissues, the measured fluorescence signals correlated morphologically with connectivetissue fibers (mainly collagen) in all layers of the bowel wall and with cytoplasmic granules within

©2003 CRC Press LLC

Page 22: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

eosinophils present between the crypts in the lamina propria of the mucosa. In addition, fluorescentsignals from the crypts of normal cells were very faint. However, a significant fluorescence emission wasobserved in the cytoplasms of dysplastic epithelial cells in the crypts of colonic adenomas. In the laminapropria of colonic adenomas, a decrease in fluorescence intensity compared to that of normal colon tissuewas found to exist. The decrease could be correlated to fewer fluorescent connective tissue fibers in thecase of the adenomas. Finally, it was found that a larger number of fluorescent eosinophils exist inadenomatous colonic tissue than in normal colon tissue.68 Similar results were also found in anotherstudy, in which a confocal fluorescence microscope was used.69

In an additional study, also aimed at determining the biochemical differences between fluorescencesignals from normal and adenomatous colonic tissues, it was found that the fluorescent signals fromplasma-soluble melanins derived from various sources were related to the overall autofluorescence signalof adenomatous tissue samples. It was also determined in this study that the fluorescence component ofmelanins derived from 3-hydroxyanthranilic acid, excited by 324-nm light and measured at 413 nm, wasless than the fluorescence caused by melanins derived from other sources (i.e., dopa, catecholamines,catechol, and 3-hydroxykynurenine), with fluorescence excitation and emission maxima at 345 and 445nm, respectively. 70

In a quantitative determination of the efficacy of fluorescence spectroscopy to classify colonic tissuesas adenoma, adenocarcinoma, or non-neoplastic, a series of analyses were performed on 83 biopsyspecimens from several patients. In these tests, fluorescence emission spectra covering a spectral rangeof 450 to 800 nm were measured. From these measurements, it was found that the spectral propertieswere significantly different for the three different tissue classifications. In fact, the results of these analyseshad a sensitivity of 80.6 and 88.2%, and a specificity of 90.5 and 95.2% in discriminating neoplastic fromnon-neoplastic mucosa and adenoma from non-neoplastic mucosa, respectively.71

With preliminary fluorescence analyses of colon tissues providing promise for a minimally invasivereal-time classification procedure for colon cancer, a great deal of effort is being devoted toward theoptimization of this procedure, both experimentally and through data analysis methods. In one suchstudy, fluorescence EEMs were obtained to determine the optimal excitation regions for obtainingfluorescence emission spectra that can be used to differentiate normal and pathologic tissues. In the caseof normal and adenomatous colon tissue, the optimal excitation wavelengths were found to be 330, 370,and 430 nm ± 10 nm. These excitation wavelengths were used with simple difference techniques as wellas ratiometric analyses to determine the optimal emission wavelengths.

Based upon these results, the optimal excitation wavelength for discrimination between normal andadenomatous colon tissues in vitro was found to be 370 nm and the optimal emission wavelengths foranalysis based upon this excitation were found to be 404, 480, and 680 nm.72 In a second study, in whichthe optimization was based upon data analysis algorithms, 35 resected colonic tissue samples were excitedby the 325-nm line of a helium cadmium laser and the resulting fluorescence spectra were measured overthe spectral range of 350 to 600 nm. Scores were derived from these analyses by a multivariate linearregression analysis that accounted for six wavelengths in the emission spectra. Normal tissue samples,adenomatous tissue samples, and hyperplastic tissue samples were classified by using the resulting scoreswith accuracies of 100, 100, and 94, respectively. 73

Gastric Cancer — Application of fluorescence spectroscopy to in vitro detection of gastric cancers has alsobeen performed. In this work, fluorescence was measured following tissue excitation with the 325-nmlight from a helium cadmium laser.63 Fluorescence images of 72 surface areas of 21 resected tissue sampleswere recorded in six regions of the visible spectrum by a cooled charge-coupled device (CCD) camera.The fluorescence emission intensities measured at 440 and 395 nm, normalized to the intensity measuredat 590 nm, differed significantly for malignant tissues, premaligant tissues, and normal gastric tissues.When these differences were used as a diagnostic parameter, classification of malignant tumor tissueswith a sensitivity of 96% and a predictive value of 42% was possible. Additionally, the same approachapplied to diagnosis of abnormal (but nontumor) stomach tissues gave values of 80 and 98%, respectively.74

©2003 CRC Press LLC

Page 23: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

Oral Cancer — Biopsy specimens from clinically suspicious lesions and normal-appearing oral mucosawere obtained from patients and fluorescence EEMs were measured.56 From these EEMs, it was determinedthat the optimal excitation wavelength for differentiation between normal and dysplastic tissues was 410nm. Based upon the fluorescence emission spectra produced by this wavelength for excitation, the 22different resected samples, 12 histologically normal and 10 abnormal (dysplastic or malignant), wereanalyzed. From this analysis, 20 of the 22 samples were correctly classified as normal or abnormal tissues.63

Atherosclerosis — Laser-induced fluorescence has been used for the differentiation of calcified and non-calcified plaques in arterial tissues.64 In this work, 248-nm laser light from a krypton-fluorine excimerlaser was used to irradiate normal and severely atherosclerotic segments of human postmortem femoralarteries. In order to analyze deeper layers of the tissue while measuring the resulting fluorescence fromeach layer, 16-ns laser pulses were used, each having a fluence of 5 J/cm2. Pulse powers of this magnitudewere sufficient to ablate as well as excite the tissue. By synchronizing the detector with each of the laserpulses, it was possible to determine the characteristic composition of the ablated tissue layer. Normaltissue layers provided fluorescence spectra exhibiting a broad-continuum emission between 300 and 700nm with peak fluorescence of equal intensity at wavelengths of 370 to 460 nm. Fluorescence maxima ofatheromas without calcification occurred at the same wavelengths but with significantly reduced intensityat 460 nm.

In contrast to these broad-continuum fluorescence signals from normal and noncalcified atheromas,calcified plaques displayed multiple-line atomic fluorescence (atomic emission) with the most prominentpeaks at wavelengths of 397, 442, 450, 461, 528, and 558 nm. These fluorescence/emission criteriaidentified the histologically classified target tissue precisely. Comparison of this technique to histologicalexamination of the corresponding arterial layers indicated an extremely accurate characterization. Theseresults demonstrated the potential for simultaneous real-time tissue identification and the feasibility ofablation by laser irradiation under strict laboratory conditions.75

To diagnose arterial tissue before ablation, many researchers have attempted to use less powerful andless energetic lasers for fluorescence excitation alone, and have experienced a great deal of success. Inone such study, ultraviolet excited laser-induced fluorescence (LIF) using excitation wavelengths rangingfrom 306 to 310 nm was used to distinguish between normal and atherosclerotic arteries.65 In this study,two distinct fluorescence emission bands were observed in the LIF spectra of normal and pathologicaorta: a short-wavelength band peaking at 340 nm (attributed to tryptophan) and a long-wavelengthband peaking at 380 nm (attributed to a combination of collagen and elastin). In addition, the intensityof the short-wavelength band was found to be very sensitive to the choice of excitation wavelength whilethe long-wavelength band remained unchanged; this feature allowed for the relative contributions ofeach band to be controlled precisely by choice of excitation wavelength.

It was found that, by using 308-nm excitation to observe emissions from the short- and long-wave-length bands simultaneously, two ratios could be determined; from them it was possible to distinguishbetween normal and atherosclerotic aortic tissue. These two ratios characterized the relative tryptophanfluorescence content and the ratio of elastin to collagen. In addition, normal and atherosclerotic aortawere correctly distinguished in 56 of 60 total cases by a binary classification scheme in which theseparameters were combined. Furthermore, atherosclerotic plaques, atheromatous plaques, and exposedcalcifications could be classified individually with sensitivities and predictive values of 90 and 90%, 100and 75%, and 82 and 82%, respectively.76

In another study, even less energetic light was used to excite autofluorescent species in blood vesselsfor the differentiation of normal tissue and atherosclerotic lesions. The use of longer wavelengths of lightfor excitation not only caused less photodamage to the irradiated tissue, but also allowed better trans-mission through optical fibers, which were necessary for in vivo light delivery. In this work, 325-nm lightwas used for excitation of the sample, and a ratio of the fluorescence intensity at 480 nm to the fluorescenceintensity at 420 nm was determined for each of the different types of tissues. Based upon these ratios, aclear differentiation between normal and mild, as well as between normal and severe, atheroscleroticlesions was observed. In the case of normal tissue, there was an increased intensity in the range from

©2003 CRC Press LLC

Page 24: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

420 to 540 nm, whereas atherosclerotic lesions had no or only a small peak at 480 nm. In addition, thespectroscopic results showed no differences between samples taken from different types of vessels.77

Further studies have extended the wavelengths used for excitation of atherosclerotic tissue samples to458 nm78 and even 476 nm.79 In both cases, even though the autofluorescent tissue components respon-sible for the fluorescence emission were different from those excited by ultraviolet light, good differen-tiation between normal arterial tissue, noncalcified arterial plaques, and calcified arterial plaques wasdemonstrated. When these longer wavelengths are used for excitation, differentiation is based upon theratio of the fluorescence signals from structural proteins (elastin and collagen) and ceroid, which can becorrelated to the concentration of these various components.

Several other studies have been performed to correlate LIF signals to the histochemical compositionof human arteries and veins.80,81 In one such study, unstained frozen sections of normal and atherosclerotichuman aorta and coronary artery were examined by histochemical and fluorescence microscopy tech-niques to identify the species responsible for autofluorescence following excitation with light ranging from351 to 364 nm. The species investigated included the structural proteins elastin and collagen in normaland atherosclerotic specimens, calcium deposits in calcified plaques, and granular or ring-shaped depositshistochemically identified as ceroid found in calcified and noncalcified plaques. The emission wavelengthand intensity of ceroid autofluorescence differed greatly from those of elastin or collagen, with the ceroidemission red-shifted and showing a much greater resistance to photobleaching than the structural proteins,thus making it a prime candidate for investigation in the classification of arterial plaques.80

In another study aimed at determining the relationship between the histochemical and morphologicalcharacteristics of atherosclerotic tissue and their LIF spectra, unstained frozen sections of 47 normal andatherosclerotic human aortas and coronary arteries were excited by 476-nm laser light. The resultingfluorescence images were measured with a bright-field epifluorescence microscope. The samples werethen stained with various dyes (including H&E, Movat pentachrome, and oil red O) for histologicalanalysis. In the case of normal artery autofluorescence, the signals correlated with the structural proteinselastin and collagen in the intima, media, and adventitia. In atherosclerotic plaque, autofluorescencecorrelated morphologically with lipid or calcific deposits in the atheroma core. The autofluorescence ofthese deposits was different from that of elastin and collagen in distribution, intensity, and wavelength,and increased with the severity of the plaque.

Such excellent correlation qualitatively and quantitatively suggests that 476-nm excitation-based LIFanalyses of arterial tissues would be useful.81 However, it has recently been found that 476-nm-inducedautofluorescence in arterial tissues suffers from changes in two prominent spectral characteristics of theemission spectrum. The changes related to alterations in the individual tissue chromophores being excitedare a permanent decrease in the peak fluorescence intensity and a reversible change in the fluorescenceemission profile (line shape). The permanent changes in absolute fluorescence intensity are due toirreversible photodamage to the tissue fluorophores; the reversible changes in fluorescence line shape aredue to some type of alteration in tissue absorbers. An attempt has been made to minimize these effects;excitation intensity levels and exposure times have been investigated to establish the thresholds at whichthese alterations are minimized.82,83

To develop a fluorescence guidance system for the laser ablation of arterial plaques in femoral arteries,Gaffney and co-workers developed a technique that employs dual excitation wavelengths. Informationabout the fatty plaque content could be obtained from 325-nm excited fluorescence; information aboutstructural proteins and calcific content could be obtained from fluorescence following 476-nm excitation.The fluorescence emission spectra obtained from each of the excitation wavelengths were used to performratiometric analyses. For 325-nm excited fluorescence spectra, 78 ratios were determined based upon13 different wavelengths, and for 476-nm excited spectra, 55 ratios were determined based upon 11different wavelengths. From these analyses, atherosclerotic lesions in human coronary arteries werecharacterized by an increase in normalized fluorescence intensity at longer wavelengths when excitedwith either ultraviolet or visible light. Calcific plaque content greater than 10% in lesions more than 1-mm thick were identified by increased normalized fluorescence intensity at 443 nm produced by exci-tation at 325 nm. Fatty plaque content was found to correlate with fluorescence intensity ratios produced

©2003 CRC Press LLC

Page 25: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

by 325-nm excitation, whereas fibrous and calcific content correlated well with fluorescence ratios during458-nm excitation.78

In addition to using multiple-excitation wavelengths to increase the accuracy and sensitivity of predic-tion of atherosclerotic tissue, there has also been a recent investigation into the use of time-resolvedfluorescence detection rather than spectral-based detection for the differentiation of atherosclerotic tissuefrom normal tissue. Based upon the temporal differences in the fluorescence emission from atheroscleroticplaques and normal blood vessels, an enhanced differentiation between the two tissue types was reported.84

Heart Arrhythmia — Autofluorescence spectral properties of tissue have also been used in the diagnosisof heart-related illnesses. Heart arrythmias are often caused by formation of nodal conductive tissues;in severe cases that cannot be treated through conventional medical therapies, transcatheter ablation ofthe nodal conductive tissue can be performed. Therefore, prior to any tissue ablation, it is important tobe able to classify the tissue of interest as nodal conductive tissue, atrial endocardium, or ventricularendocardium. To perform this diagnosis, Lucas and co-workers studied the fluorescence emission ofin vitro heart tissue samples excited by the 308-nm laser line of an XeCl excimer laser (1.5 mJ/pulse, 10Hz). Following excitation at 308 nm, the nodal tissue could clearly be distinguished from atrial endomy-ocardial tissue by a visible decrease in fluorescence emission intensity between 440 and 500 nm, peakarea between 440 and 500 nm, and peak width. Nodal conduction tissue could also be distinguishedfrom ventricular endocardium by its relative increase in fluorescence emission between 430 and 550 nm.Specificities of 73 and 88% and sensitivities of 73 and 60% were possible for sinus nodal and atrioven-tricular nodal conduction tissue identification, respectively.85

Tooth Decay — The field of dentistry has also seen a recent growth of research in the use of fluorescencespectroscopy. In work involving the use of a confocal scanning laser microscope (CLSM), autofluorescencespectra were taken of teeth with demineralized dentin on the root surfaces. The resulting spectra werethen compared with spectra taken at other locations on the tooth with minimal to no demineralizationin the dentin. When observed in CLSM images, demineralized dentin (excited at 488 nm) exhibited anincreased fluorescence emission at 529 nm when compared with the spectra of healthy dentin. Thisdifference in fluorescence intensity decreased deeper into the root, as the healthy dentin underneath thelesion was beginning to be excited.

In contrast, when fluorescence spectrophotometry was used with excitation around 460 and 488 nm,it yielded fluorescence emission intensity (about 520 nm) for demineralized dentin that was lower thanfor healthy dentin, but in a more pronounced peak. From excitation spectra obtained by a fixed emissionwavelength of 520 nm, it could be seen that the contribution of excitation between 480 and 520 nm wasmore important in demineralized dentin than in healthy dentin. Because of the small sampling volumeused in CLSM image acquisition, the recorded fluorescence was not affected by demineralization-inducedchanges in scattering and absorption properties. Thus the increased fluorescence for demineralized dentinimplies an increased quantum yield. However, in fluorescence spectrophotometry, where the measure-ment volume is large relative to the lesions, changes in scattering and absorption properties do have aninfluence on the fluorescence signal. Therefore, increased absorption by nonfluorescing chromophoresand increased reabsorption around the emission wavelength may compensate for the increase in quantumyield and absorption around the excitation wavelength by the fluorophores.86

Eosinophils — Eosinophils are rare granulocytes typically associated with allergic diseases or responsesto various parasitic infections. Many types of human cancers, however, are also associated with extensiveeosinophilia, either within the tumor, in the peripheral blood, or in both locations. Special techniquessuch as autofluorescence or immunohistochemistry are sometimes needed to detect the presence of intactand degranulating eosinophils within the tumors. With the help of these techniques, extensive amountsof eosinophilia have been found in hematologic tumors such as Hodgkin’s disease and certain lymphomas.However, many other types of cancer, such as cancers of the colon, cervix, lung, breast, and ovary, alsocontain eosinophilia, and it can be identified if diligently sought. Although the presence or absence ofeosinophilia within these tumors does not appear to have a major influence on the prognosis of the

©2003 CRC Press LLC

Page 26: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

disease, eosinophils may play an important role in the host interaction with the tumor, perhaps bypromoting angiogenesis and connective tissue formation adjacent to the cancer.

In addition, tumor-related eosinophilia provides some interesting clues into tumor biology, particularlywith regard to production of cytokines by the tumor cells.87 Characterization of the fluorescence propertiesof human eosinophils isolated from peripheral blood of normal donors has been performed by measuringEEMs over a wide range of wavelengths. Circulating eosinophils possess three fluorescence emissionmaxima: one at 330 nm following excitation at 280 nm, which can be attributed to tryptophan, a secondpeak at 440 nm following excitation at 360 nm, and the last at 415 nm following excitation with 380-nmlight. Fluorescence microscopy studies also showed that the fluorescence of eosinophils appears to be sitedependent. For instance, when observed following excitation by 365-nm light, circulating eosinophilsfluoresce blue–violet, while tissue-dwelling eosinophils fluoresce amber–gold. Therefore, when fluores-cence spectroscopy is used to develop optical biopsy techniques based upon eosinophils in human tissue,the differences in their local environments may have a significant impact on fluorescence spectra.88

28.4.2.2.2 Tissue Analysis Using Exogenous DyesDue to the significant differences in tissue uptake and storage of various exogenous fluorophores betweenin vitro and in vivo specimens, relatively few studies have been performed in this area in terms ofdeveloping diagnostic procedures. In most cases where exogenous fluorophores are used, the actuallocation and kinetics of tissue uptake are important; therefore, because in vitro tissues differ in theseproperties, such studies generally do not provide any useful information. However, properties such astissue reactivity to a specific chemical can be studied by using in vitro systems to provide a betterunderstanding of the mechanism by which specific reactions take place.

Lipid Peroxidation — One system studied by using fluorescence CLSM is the well-established experi-mental model of lipid peroxidation induced by haloalkane intoxication in liver tissues. In this study, thefluorescent reagent 3-hydroxy-2-naphtholic acid hydrazide was used to derivatize the carbonyl functionalgroups originating from the lipoperoxidative process in liver cyrostat sections from in vivo intoxicatedrats, as well as isolated hepatocytes that were exposed in vitro to the haloalkanes. The resulting CLSMimages were able to visualize the tissue areas and the subcellular sites first involved in oxidative stressand lipid peroxidation. The images also showed that haloalkane-induced lipid peroxidation in hepato-cytes primarily involves the perinuclear endoplasmic reticulum, whereas the plasma membrane and thenuclear compartment are unaffected, and that lipid peroxidation also induces an increase of liverautofluorescence.89

28.4.3 In Vivo Analyses and Diagnostics

While in vitro studies can reduce the complexity of the biological system analyzed and provide usefulinformation about basic biological functions or reactions, the need to develop in vivo analysis techniquesfor medical diagnosis of diseases is critical. The range of fluorescence-based in vivo diagnostic techniquesspans from monitoring of atherosclerosis90 to the detection of tooth decay.60,91 One of the most commonbiomedical diagnostic procedures performed using fluorescence spectroscopy is known as “opticalbiopsy.”10,24,92–102 In this procedure, some form of optical spectroscopy, typically fluorescence spectroscopy,is used to identify differences between healthy, malignant, and premalignant tissues of various organs.Over the past two decades, a great deal of research has been performed in this field for the diagnosis ofmany different forms of cancer.6,59,61,65,99,103–120 In order to ensure that the various diagnostic procedureswill work in complex living systems, two types of analyses are generally performed: animal studies andclinical human trials.

28.4.3.1 Animal Studies

Animals typically provide an excellent test system for many different types of disease diagnoses prior toclinical studies. The key in choosing an animal for these studies is to ensure that the biochemical structureof the particular organ or location of the animal used is comparable to that of a human. In addition, the

©2003 CRC Press LLC

Page 27: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

animal should be relatively easy to care for and should match any other criterion for the particular typeof analysis to be performed. Mice and rats are often used as a suitable alternative for humans in manypreliminary studies because of their 98% genetic compatibility with humans and because they arerelatively easy to handle; however, many other animal models have also been investigated (e.g., hamsterand pigs).

In the case of “optical biopsy” work or photodynamic therapy (PDT), transgenic mice with trans-plantable tumors are the most common model system. These mice have been genetically altered to preventtheir providing an immune response to the tumors that are induced in them. By using these mice withtransplantable tumors, valuable information useful to later clinical trials can be obtained. However, useof animal models could have artifacts as well. For instance, when using mice with transplantable tumorsfor tumor detection diagnostics, the implanted tumor tissue generally comes from a different type oftissue and therefore may not have the same biochemistry, architecture, and vasculature as the tissue intowhich it is being placed.

In addition, unlike spontaneous tumors in humans, transplanted tumors often remain mostly sepa-rated from the normal tissue. When exogenous fluorophores are used as contrast agents in fluorescencediagnostics, there is also an issue of species-dependent chemical distribution or pharmacokinetics. Theseproblems also begin to become convoluted by the fact that drug pharmacokinetics are often very differentbetween early stages and more advanced stages of cancer or transplanted tumors because vascularizationdiffers.121,122 One way to minimize these problems in tumor studies is to induce the tumor chemically orradiologically. This will allow the tumor to be integrated into the tissue as well as ensure that the tumorbeing investigated comes from the same type of tissue in which it is growing.

One well-established tumor model procedure is to use dimethyl benzanthracene (DMBA) to inducelesions in the cheek pouches of hamsters. This particular model is useful because it progresses throughmany different stages of cancer development: normal tissue to hyperplastic tissue to dysplastic tissue tocarcinoma in situ (CIS), and finally to invasive carcinoma.123 Endogenous and exogenous fluorophoreshave been used for in vivo diagnostics based on this model. In the case of autofluorescence diagnostics,it has been reported that 76% sensitivity and 86% specificity have been obtained in the detection of earlyneoplastic tissue. In addition, by intravenously administering the photosensitizer Photofrin™ 24 h priorto analysis, the sensitivity and the specificity could be increased to 100%.

28.4.3.1.1 Animal Studies Using AutofluorescenceWith analyses on living animals, it is possible to gain a much more accurate picture in most cases thancan be achieved in vitro. For instance, one of the strongest chromophores present in tissues is hemoglobin(oxy- and deoxy-); in in vitro studies, no blood flow is present, and the ratio of oxygenated hemoglobinto deoxygenated hemoglobin is very different from that in live animal studies. This factor alone couldhave a significant effect on the autofluorescence spectrum of the tissue by absorbing the fluorescent lightor by changing the oxidative state of some of the autofluorescent chemicals present.

Cancer Diagnostics — Esophageal Cancer — The capacity to identify subclinical neoplastic diseases ofthe upper aerodigestive tract using tissue autofluorescence spectroscopy has significantly contributed tothe field of fluorescence cancer screening. In 1993, the applicability of tissue autofluorescence for theearly diagnosis of precancerous states in esophageal mucosa was studied through various model sys-tems.124 In an N-nitroso-N-methylbenzylamine (NMBA)-induced rat esophageal cancer model, alter-ation of the fluorescence emission pattern at 380 nm was found to correspond to disease progressionfrom normal mucosa through dysplasia to invasive cancer. Although gross assessment of the tissue wasindistinguishable from the saline-treated controls, histopathologic evaluation revealed NMBA-inducedpreneoplastic changes in the epithelium.125 In addition, a multicellular tumor spheroid model, inducedby transretinoic acid (RA), was also found to alter autofluorescence intensities at multiple wavelengthsincluding 340, 450, and 520 nm. Such RA-induced alterations corresponded to changes in the state ofspheroid differentiation.124

©2003 CRC Press LLC

Page 28: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

Brain Cancer — Brain tumors are one of the more difficult types of cancer to treat. Unlike many otherforms, where tumor margining can be less important and healthy tissue surrounding the tumor can beremoved to ensure that no malignant cells are left behind, the removal of excess brain tissue during tumorremoval can have dramatic adverse effects. Because of this important requirement, a technique capableof providing an accurate demarcation between malignant cells and normal brain cells is extremelyimportant. One method that is being developed is autofluorescence-based optical biopsy.

In initial animal studies, EEMs of rat gliomas revealed three distinct regions of decreased autofluores-cence emission with respect to the normal rat brain tissue. These differences in the fluorescence emissionspectra of the two different types of tissue samples occurred at 470, 520, and 630 nm, with correspondingexcitation wavelengths of 360, 440, and 490 nm. The fluorescence emission at 470 nm corresponds toNAD(P)H, while the emissions of 520 nm and 630 nm correspond to various flavins and porphyrins,respectively. Due to the nature of the chemical differences between the normal tissue and the gliomatissue, this finding suggests a relationship between brain tissue autofluorescence and metabolic activity.This is in contrast to in vitro brain tissue studies, which also found that NAD(P)H fluorescence was lowerin all measured human brain tumors, but, depending on their nature, flavin and porphyrin autofluores-cence in neoplastic tissues was not always lower than in normal tissue.126

Atherosclerosis — Due to the high occurrence rate of atheriosclerosis in adults and the preliminary successof LIF in diagnosing the disease in its various stages in vitro, animal studies have been performed toensure that the same fluorescence phenomena occur in living systems. In one such study, an XeCl excimerlaser operating at 308 nm was used to excite proteins of human aortas containing early lipid-richnoncollagenous lesions. The emitted fluorescence exhibited significant red shifts and spectral broadeningcompared with spectra from nonatherosclerotic human aortas. Similar red-shifted and spectrally broad-ened autofluorescence profiles were observed from oxidatively modified low-density lipoproteins excitedby 308-nm illumination. However, in the case of native low-density lipoproteins, neither the red shiftnor the spectral broadening was found to exist. In order to compare these tissue results to the autoflu-orescence emission from live animals, LIF studies were performed on hypercholesterolemic rabbits withearly foam cell lesions. The resulting autofluorescence spectra were similar to those of oxidized beta low-density lipoprotein, the major lipoprotein accumulating in arteries of rabbits fed cholesterol and an earlyindicator of atherosclerosis.127

28.4.3.1.2 Animal Studies Using Exogenous DyesThe study of exogenous fluorophores in living animals can provide a great deal of information. In additionto being a model system that more closely resembles human when testing fluorescence-based diagnosticsystems prior to clinical trials, they also play a large part in the field of pharmacokinetics. Using livinganimals with functioning circulatory systems, it is possible to watch the real-time distribution of variousdrugs throughout the body as well as study their uptake into various tissues and, finally, their excretionfrom the body. This is an extremely important field of research because it ensures timely deliverey ofdrugs to the target tissue areas of interest.

Cancer Diagnostics — Some of the more commonly studied exogenous fluorophores in biomedical anal-yses are used for cancer diagnosis or treatment, or both. These fluorophores, known as photolabels inthe case of the former and photosensitizers in the case of the latter, represent an extensive class ofcompounds whose pharmacokinetics properties as well as their tumor demarcation abilities need to bestudied prior to use in human trials. These studies are generally performed on animal subjects; severalof the more common ones are described in the following sections.

Pharmacokinetics of Photosensitizers — Photosensitizers represent a unique class of compounds that,upon being activated by the absorption of a specific wavelength of light, are capable of killing thesurrounding tissues. A detailed description of the mechanism of action of these photosensitizers can befound in Chapter 36 of this handbook. Therefore, only salient features are briefly described here. The

©2003 CRC Press LLC

Page 29: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

concept behind these photosensitizers is to develop drugs that are preferentially taken up or localized inmalignant tissues and can then be photoactivated. After the drug is administered and a specific amountof time has passed for optimal uptake, light is shown on the tissue area of interest and absorbed by thesensitizer. The sensitizer then kills the surrounding tumor tissue, leaving the healthy tissue undamaged.

Although the number of available photosensitizers approved for clinical use is relatively small, a largenumber of new potential candidates are continually being developed. These new agents are then comparedwith the better characterized sensitizers based upon many parameters, including tissue localization ability,effectiveness in promoting cell death, and toxicity, as well as several other parameters. In a study todetermine the most effective photosensitizer of five commonly used drugs, their tissue localizationproperties were studied in vivo using “sandwich” observation chambers and tumors that were growingin thigh muscle. Several common dyes were studied, including hematoporphyrin derivative, PhotoFrinII, aluminum phthalocyanine tetrasulphonate, uroporphyrin I, and acridine red. Of the photodynamicallyactive dyes (the first three), aluminum phthalocyanine tetrasulphonate was found to exhibit the best invivo tumor localization properties as determined by fluorescence spectroscopy.128

LIF studies have also been used to investigate the pharmacokinetic properties of 5-aminolevulinic acid(ALA)-induced protoporphyrin IX (PpIX). These analyses were performed in normal and tumor tissuesof rats following intravenous (i.v.) injection of ALA. The aim of the study was to investigate ALA-induced(PpIX) formation and its accumulation in different types of rat tissues after the systemic administrationof ALA. Tissue types investigated included a malignant rat tumor and normal tissue from 13 differentorgans in 8 rats. The various rats were injected with two different doses of ALA (30 and 90 mg/kg bodyweight), and fluorescence analyses were performed 10, 30, and 240 min after injection.

Fluorescence analyses were performed by exciting the sample with 405-nm light and monitoring theresulting fluorescence with a fiber-optic probe over the spectral region of 400 to 750 nm. The fluorescencesignal consisted of a broadband autofluorescence background with a maximum at approximately 500nm and a characteristic dual-peak emission from the PpIX at 635 and 705 nm. From this work, it wasfound that the maximum tumor buildup of PpIX was achieved in less than 1 h after ALA injection, thatthe fluorescence demarcation between tumor and surrounding tissue was between 7:1 and 8:1 after 30min, and that it decreased with longer retention times. Of the 13 different organs investigated in thisstudy, PpIX buildup was found to be particularly high in the stomach and the intestine.129

Another class of exogenous dyes that have been used for tumor demarcation are hematoporphyrinsand hemotoporphyrin derivatives. In a pharmacokinetic study of hematoporphyrin, rats were adminis-tered i.v. injections of the dye, followed by immediate and continuous analysis by LIF. Excitation of thehematoporphyrins was provided by the 337-nm output of a nitrogen laser; the signal was collected byan optical multichannel analyzer (OMA), which allowed for acquisition of entire fluorescence spectrumfor each laser shot. Upon analysis of data from several of the rat’s organs, it was found that the fluorescenceemission from the hematoporphyrin at 630 nm exhibited an initial peak intensity as well as a delayedpeak intensity. The delayed fluorescence peak was described as due to the chemical components ofintracellularly transformed hematoporphyrin derivatives. In addition, it was also discovered that, bydividing the background-free 630-nm signal by the autofluorescence intensity at shorter (blue) wave-lengths, a ratio exhibiting a larger contrast between tumor and surrounding tissue could be obtainedthat would greatly aid in tumor demarcation.130

A third and relatively recent photosensitizer used for the demarcation of tumor tissues is benzopor-phyrin derivatized-monoacid (BPD-MA). In LIF studies designed to provide pharmacokinetic informa-tion about this compound, 337-nm light from a nitrogen laser was used to excite the BPD-MA in ratsthat had been administered the drug via i.v. The fluorescence emission, over the spectral range of 380 to750 nm, was then monitored with a spectrometer equipped with a diode array detector. Three hoursafter the injection period, the fluorescence signals were measured from many different types of rat tissue,including malignant tumors that had been experimentally induced. These results were then comparedwith results from several other common sensitizers, such as hematoporphyrin (HP), polyhematoporphy-rin ester (PHE), tetrasulfonated phthalocyanine (TSPc), and the commercially available Photofrin™. Afterthree hours, the demarcation potential between tumor and surrounding tissue in terms of fluorescence

©2003 CRC Press LLC

Page 30: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

signal for the tumor model used was 2:1 for BPD-MA. When compared with other drugs such as HP, itshows about the same demarcation potential, whereas Photofrin and PHE exhibit about 3 times betterand TSPc about 1.5 times better demarcation. Employing the endogenous tissue fluorescence signatureenhanced the contrast by a factor of about two for each of the five drugs.131

In work by Nilsson et al., the biodistribution of two recently developed tumor markers, trimethylated(CP[Me]3) and trimethoxylated (CP[OMe]3) carotenoporphyrin, was investigated by means of LIF. Inthis study, 38 tumor-bearing (MS-2 fibrosarcoma) female BALB/c mice were administered the drugsthrough i.v. injection. At 3, 24, 48, or 96 h after administration, the carotenoporphyrin fluorescence wasmeasured in tumoral and peritumoral tissue, as well as in the abdominal, thoracic, and cranial cavities.Excitation of the exogenous dye was performed by using the 425-nm emission of a nitrogen-pumpeddye laser and was measured by a spectrometer equipped with an intensified charge-coupled device (ICCD)at 490, 655, and 720 nm. The emission bands at 655 and 720 nm corresponded to carotenoporphyrin(CP), whereas the band at 490 nm represented a location near the maximum of the autofluorescenceemission of the tissue. The tissues that showed the greatest extent of CP-related fluorescence were thetumors and the liver tissues, whereas the cerebral cortex and muscle tissues consistently exhibited weakCP-related fluorescence. Additionally, although the fluorescence intensity of most tissue types decreasedover time, it was found that the fluorescence intensity in the liver remained constant the full 96 h overwhich it was investigated.132

Oral Cancer — Apart from pharmacokinetic analyses of various sensitizers, animal studies with thesecompounds have also been performed to verify and quantify the ability of these drugs for tumor demar-cation in different types of tissues. One such study investigated the effectiveness of porfimer sodium-derived drugs for the detection of early neoplastic changes in the oral cavity of hamsters. Neoplasia wasinduced in the hampsters’ cheek pouches by the application of 9,10-dimethyl-1,2-benzanthracene. Fol-lowing formation of neoplastic tissue in the hamster cheek pouch, autofluorescence analyses as well asfluorescence analyses using porfimer sodium as an exogenous tumor demarcation drug were performedand compared with histologic results of the same tissue sites. When the two fluorescence diagnostictechniques were compared with the histological results, it was found that the autofluorescence analysiswas capable of 76% sensitivity and 83% specificity, whereas the porfimer sodium-based fluorescencetechnique provided 100% sensitivity and specificity for the samples investigated.133

Pancreatic Cancer — Pheophorbide-a (Ph-a) represents another of the many different types of tumordemarcation drugs that have been used for optical differentiation of normal tissues and tumor tissues.To test its viability for the detection pancreatic tumors, LIF analyses of Ph-a were used to image sixintrapancreatic tumors and six healthy pancreases in vivo in rats. Ph-a was intravenously administeredto the rats at a concentration of 9 mg/kg body weight, and fluorescence images were acquired up to 48h after injection of the drug. Excitation of the blue tissue autofluorescence was performed by using 355-nm light from a frequency-tripled Nd:YAG laser and excitation of the dye was performed by the 610-nmoutput from an Nd:YAG pumped dye laser. Fluorescence images were obtained at three different wave-lengths; bandpass filters were used for wavelength selection. Images at 470 nm and 640 nm were used tomonitor the autofluorescence of the tissue, while images at 680 nm were taken to obtain compositeimages of the dye and autofluorescence emissions together. In order to achieve a good contrast betweenthe normal pancreatic tissue and the tumor tissue, the autofluorescence intensity of the 640-nm imagewas normalized to the background fluorescence intensity in the 680-nm image prior to being subtractedfrom the 680-nm image. Following this subtraction, the differential image was divided by the autofluo-rescence image at 470 nm. The resulting ratiometric images allowed for safe diagnoses to be made byproviding well-contrasted tumor images.24

Brain Tumors — Animal models have also been used in studies in which hematoporphyrin derivative(HPD) was used for the diagnosis of brain tumors. In this study, adult Wistar rats had C6 glioma cellsimplanted into their brains to act as a cerebral glioma model. After the tumors had developed to a sizeof 7 to 12 mm in diameter, they were injected intravenously with 5 mg/kg body weight of HPD, and

©2003 CRC Press LLC

Page 31: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

continuous fluorescence analyses were performed for 24 h using a fiber optic-based fluorimeter. Thefluorescence intensity of the normal brain tissue reached a plateau 60 min after injection, while the gliomaregion reached a plateau 80 min after injection. Fluorescence analyses of glioma, brain tissue adjacent tothe tumor (BTAT), and normal brain tissue 24 h after injection revealed that the fluorescence intensityof the glioma was 6.1 times greater than that of normal brain tissue; the BTAT also was 3.9 times greaterthan the normal tissue.134

Liver Cancer — Testing efficacy of PDT using ALA-induced PpIX sensitization for the treatment of heptictumors was performed with rat models. Liver tumors were induced in the rats by local innoculation oftumors cells or by administration of the tumor cells through the portal vein. Following tumor formation,60 mg/kg body weight of ALA was administered intravenously. After 60 min, the PpIX in the heptictumor tissue was excited, initiating the PDT treatment as well as allowing the PpIX to be excited. Theresulting fluorescence emission was monitored at 635 nm. Fluorescence analyses revealed that largeaccumulations of the PpIX occurred in the heptic tumor, as well as in normal liver tissue, but not in theabdominal wall muscles.

In addition to fluorescence analyses, laser Doppler imaging was used to determine changes in thesuperficial blood flow in connection with PDT. Histopathological examinations were also performed toevaluate the photodynamic therapy effects on the tumor and the surrounding liver tissue, includingpathological features in the microvascular system. Laser Doppler imaging results indicated an effect onthe vascular system in the tumor as well as on the surrounding tissue following PDT treatment, asdetermined by decreased blood flow in the treated area. In addition, the tumor growth rate decreasedsignificantly when evaluated 3 and 6 days after the treatment, showing that ALA-induced PDT holdspromise for treatment of heptic tumors.135

28.4.3.2 Human Studies and Clinical Diagnostics

By far the most common type of fluorescence-based biomedical diagnostic procedure used in clinicalstudies is the optical detection of malignant or premalignant tissues in various organs. Among the morecommon types of tumors that have been investigated with this technique are skin, urinary bladder,bronchus, gastrointestinal, head and neck, gynocological, breast, and brain cancers. Currently, two verydifferent approaches to these fluorescence-based optical biopsy techniques are employed. The first ofthese techniques relies on subtle differences in the tissue composition and morphology between normal,dysplastic, and malignant tissues and their effect on the autofluorescence properties for differentiation.Examples of this type of diagnoses are discussed in the next section of this chapter. The second type offluorescence-based diagnostic technique relies on the presence of exogenous fluorophores, such as PDTphotosensitizers, for the tissue differentiation.

28.4.3.2.1 Clinical Studies and Diagnostics Using AutofluorescenceAutofluorescence-based optical biopsy techniques represent the ideal form of fluorescence-based diag-nostic procedure. In these analyses, laser light is used to excite the naturally occurring fluorophores inthe tissue; differences in chemical composition between the various types of tissue will allow for real-time diagnoses without removal of a tissue sample or treatment with a contrast-enhancing drug. Becauseof this and the great deal of promise that initial studies have shown, research in this field is currentlyexperiencing tremendous growth.

Cancer Diagnostics (Optical Biopsy) — Cervical Cancer — Cervical cancer is the second most commonmalignancy in women worldwide and remains a significant health problem. Despite the widely usedPapanicolaou smear (Pap smear) screening procedure and costly treatments, the overall survival rateremains 40%. For these reasons, efforts to strengthen screening and prevention are needed.136 Autoflu-orescence optical biopsy techniques offer just such a screening technique with automated diagnosis inreal time and comparable sensitivity and specificity to colposcopy.137 Feld and co-workers have demon-strated the ability to distinguish between various types of tissues, in vivo, based upon multicomponentanalysis (Figure 28.6).63

©2003 CRC Press LLC

Page 32: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

Richards-Kortum and co-workers have used laser-induced fluorescence, employing 337-nm excitationto differentiate in vivo cervical intraepithelial neoplasia (CIN), non-neoplastic abnormal, and normalcervical tissues from one another.138 In this work, a colposcope was used to identify normal and abnormalsites on the cervix. These sites were then interrogated via fluorescence spectroscopy with an optical fiberprobe. Based upon results from the fluorescence analyses, two algorithms were developed for the diag-nosis of CIN. The first of these algorithms allowed for differentiation of histologically abnormal tissuesfrom colposcopically normal tissues with a sensitivity, specificity, and positive predictive value of 92, 90,and 88%, respectively. The second algorithm then allowed for the differentiation of preneoplastic andneoplastic tissues from non-neoplastic abnormal tissues with a sensitivity, specificity, and positive pre-dictive value of 87, 73, and 74%, respectively. These results found that as the tissue progresses from anormal state to an abnormal state in the same patient, it is accompanied by a decrease in the absolutefluorescence contribution of collagen, an increase in the absolute attenuation by oxyhemoglobin, andan increase in the relative contribution from reduced nicotinamide dinucleotide phosphate [NAD(P)H].Differentiation of the various tissues is then determined by the extent of each of these factors. Suchresults provide a great deal of hope to the future of in vivo fluorescence spectroscopy for the diagnosisof CIN at colposcopy.138,139

Esophageal Cancer — Esophageal cancers, like most other cancers, generally progress through a series ofstages. Barrett’s esophagus, named after the physician who first identified the condition,140 is an abnor-mality resulting from long-term acid reflux, and is associated with an increased occurrence of mucosaldysplasia and adenocarcinoma in the specialized glandular mucosa.141–143 In addition, individuals withBarrett’s esophagus have a tendency to develop cancer of the esophagus, with a 30- to 52-fold increaseover individuals without this condition.144,145 In esophageal cancer, the mucosa progresses from “normal”through low-grade dysplasia (LGD) to high-grade dysplasia (HGD), and, finally, cancer. If detected early,endoscopic treatment of dysplasia with photodynamic therapy or thermal ablation can be effective,thereby eliminating the need for surgical esophagectomy.

Vo-Dinh and co-workers have developed a laser-induced fluorescence diagnostic procedure for in vivodetection of GI cancer that uses 410-nm laser light from a nitrogen-pumped dye laser passed through afiber-optic probe to excite the tissue.104,105,119,120,146 After the tissue is excited, the resulting fluorescence

FIGURE 28.6 Study of in vivo quantitative fluorescent biomarkers of epithelial precancerous changes. This figuredemonstrates the ability to distinguish between various types of tissues, in vivo, based upon a multicomponentfluorescence analysis. (A) Demonstrates the ability to differentiate between normal ectocervical tissue (�), squamousmetaplasia sites (●), and high-grade squamous intraepithelial lesions (�), based upon this two-dimensional analysisprocedure. The dashed lines represent the boundary between the different tissues, based upon logistic regressionanalysis. (B) Demonstrates the ability to differentiate between nondysplatic tissues (�) and high-grade dysplastictissues sites (�). (Adapted from Georgakoudi, I. et al., Cancer Res., 62, 682, 2002.)

NA

D(P

)H F

luor

esce

nce

Collagen Fluorescence Collagen Fluorescence

NA

D(P

)H F

luor

esce

nce

A B

0 0

2

4 5 6 7

4

6

5

10

15

20

25

1 10 100

©2003 CRC Press LLC

Page 33: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

emission is collected by the same fiber-optic probe and is recorded on an OMA in 600 ms. Based uponthe resulting fluorescence spectra, a diagnostic technique known as differential normalized fluorescencewas employed to enhance the slight spectral differences between normal and malignant tissues.64,65 Thistechnique greatly improves the accuracy of diagnosis, compared to direct intensity measurements, becauseeach spectrum is normalized with respect to its total integrated intensity and therefore becomes inde-pendent of the intensity factor. This in turn enhances small spectral features in weak fluorescence signals,making classification much easier. The sensitivity of the DNF method in classifying normal tissue andmalignant tumors is 98%.65,119,120,146

The LIF methodology was also employed in clinical studies of over 100 patients, in which Barrett’smucosa without dysplasia was diagnosed with a specificity of 96% (208 of 216 data points) and high-grade dysplasia was diagnosed with a sensitivity of 90% (9 of 10 data points).104,105 Figure 28.7 shows theinstrument setup for GI cancer diagnostics.64 A photograph of the optical fiber inside the GI tract isshown in the insert of this figure. Figure 28.8 shows clinical data demonstrating the capability of LIF todifferentiate normal from malignant tissues.64

Bladder Cancer — Bladder cancer has also shown a significant amount of promise for clinical diagnosisby autofluorescence analyses. In one such study, a quartz fiber-optic probe, placed in the working channelof a cytoscope, was used to deliver 337-nm laser light from a nitrogen laser to the tissue area of interest.Following excitation, the resulting fluorescent light was collected by a second fiber and then spectrallydispersed before being detected by an OMA. Resulting fluorescence emission spectra were then analyzedby taking a ratio of the intensity of the fluorescent light at 385 nm to the fluorescence intensity at 455nm. This ratiometric analysis was used to analyze 114 lesions, and it was possible to differentiate clearlybetween malignant and nonmalignant bladder tissues with a sensitivity of 98%.147

In another study, the ability of laser-induced autofluorescence spectroscopy to distinguish betweenneoplastic urothelial bladder lesions and normal or nonspecific inflammatory mucosa was investigated.In this study, three different pulsed-laser wavelengths were used successively for excitation: 308 nm (XeCllaser), 337 nm (N2 laser), and 480 nm (coumarin dye laser). The excitation beam was delivered through

FIGURE 28.7 Laser-induced fluorescence instrument for in vivo gastrointestinal cancer diagnostics. This figureshows a typical fluorescence-based optical biopsy system in which a fiber optic probe is transmitted down a biopsychannel of an endoscope for point-by-point types of analyses. Insert: photograph of the optical fiber inside the GItract. (Modified from Vo-Dinh, T. et al., Appl. Spectrosc., 51, 58, 1997.)

DyeModule

NitrogenLaser

Poly-chromator

Multi-channelDetector

PC

Optics

Endoscope

BifurcatedOptical Fiber

B

©2003 CRC Press LLC

Page 34: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

FIGURE 28.8 Clinical data demonstrating the capability of LIF to differentiate normal from malignant tissues. Left: Differential normalized fluorescence (DNF) emission spectra of normaltissue (top) and malignant tissue (bottom). These spectra were obtained by normalizing the fluorescence intensity at every point in the spectrum to the overall area under the curve, thusemphasizing small differences between the two types of tissues. By subtracting the DNF spectrum of a particular sample from an average DNF normal tissue spectrum, a single index valueat each wavelength can be determined. (From Vo-Dinh, T. et al., Appl. Spectrosc., 51, 58, 1997.) Right: Plot of DNF indices at 480 nm for different tissue samples using in vivo LIF measurements(taken prior to biopsy). These tissue samples were then characterized via histology as normal (•) or as carcinoma (+). As expected, the normal tissue results are scattered about the index valueof zero while the carcinoma values are significantly lower due to a decrease in fluorescence intensity at this wavelength. The samples denoted by (*) represent points initially classified byhistology as normal but later reclassified as malignant. The sensitivity of the DNF method in classifying normal tissue and malignant tumors for GI cancer is 98%. (From Vo-Dinh, T.,Panjehpour, M., and Overholt, B.F., in Advances in Optical Biopsy and Optical Mammography, New York Academy of Sciences, New York, 1998, p. 116; Vo-Dinh, T., et al., Lasers Surg. Med.,16, 41, 1995.)

DIF

FE

RE

NT

IAL

NO

RM

ALI

ZE

D F

LUO

RE

SC

EN

CE

(D

NF

)

3.0

2.0

1.0

0.0

−1.0

−2.0

2.0

1.0

0.0

−1.0

−2.0

−3.0

NORMAL TISSUE

MALIGNANT TUMOR

WAVELENGTH (NM)

450 500 550 600 650 700

100

0

50

−50

−100

−150

−2000 10 20 30 40 50 60 70 80

Data Points

Normal + Carcinoma

Intensity (All Data × 10−5)

©2003 CRC Press LLC

Page 35: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

optical fibers placed in the working channel of a standard cystoscope, and the resulting fluorescence wasdetected using an OMA. When 337- and 480-nm excitation wavelengths were used, the overall fluores-cence intensity of bladder tumors was clearly decreased compared with normal urothelial mucosa regard-less of tumor stage and grade. However, when 308-nm excitation was employed, the shape of the tumorspectra, including CIS, was markedly different from that of normal or nonspecific inflammatorymucosa.148

Colon Cancer — Autofluorescence diagnostic procedures are used extensively to investigate colon cancers.In one such study, 337-nm light from a nitrogen laser was launched into a fiber-optic probe placed inthe working channel of a colonoscope and was used for excitation of in vivo and in vitro colon tissues.The fluorescence spectra were then measured and analyzed. In all cases, the spectra exhibited peaks at390 and 460 nm, which are believed to arise from collagen and NADH, as well as a minimum at 425nm, consistent with absorption attributable to hemoglobin. Despite the presence of these bands in thein vivo and in vitro samples, the relative intensity of each was quite different, especially for the NADHcomponent, whose intensity was found to decay exponentially with time after resection. Differentiationof normal colonic tissue from hyperplastic or adenomatous tissues of the same type (i.e., in vivo vs.in vitro) could be accomplished based upon a decrease in the collagen component of the autofluorescenceand an increase in hemoglobin reabsorption.

When multivariate linear regression (MVLR) analysis based upon these differences was used, neoplastictissues could be distinguished from non-neoplastic tissues with a sensitivity, specificity, positive predictivevalue, and negative predictive value toward neoplastic tissue of 80, 92, 82, and 91%, respectively. However,when this same MVLR technique was used to distinguish neoplastic polyps from non-neoplastic polyps,values of 86, 77, 86, and 77%, respectively, were obtained. This suggested that the LIF measurementssense changes in polyp morphology rather than changes in fluorophores specific to polyps, and that thischange in morphology leads indirectly to polyp discrimination.149

Laser-induced autofluorescence spectroscopy has been investigated to detect colonic dysplasia in vivousing an excitation wavelength of 370 nm.141 In this work, fluorescence emission data were used to devisean algorithm to classify colonic tissue as normal, hyperplastic, or adenomatous, based on probabilitydistributions of the fluorescence intensity at 460 nm and the ratio of the intensity at 680 nm to that at600 nm. Fluorescence spectra were then collected from normal mucosa and colonic polyps during routinecolonoscopy exams; the predictive abilities of the diagnostic algorithm were tested in a blinded fashion,with histology results the standard against which they were measured. Results revealed that the algorithmcorrectly determined the tissue type in 88% of cases, equal to the agreement of independent pathologists.In addition, the sensitivity, specificity, and positive predictive value for the detection of dysplasia were90, 95, and 90%, respectively.150

In additional work by Feld and co-workers, LIF studies were performed on 20 different patients,providing analyses on 31 colonic adenomas, 4 hyperplastic polyps, and 32 samples of normal mucosa.From these studies, it was found that classification of the tissue as adenomatous or normal colonicmucosa/hyperplastic polyp was correct, based upon an automated probabilistic algorithm, 97% of thetime. The sensitivity, specificity, and positive predictive value of the technique were 100, 97, and 94%,respectively — once again showing a great deal of promise for this technique in the future.151

Lung Cancer — Lung cancer accounts for 25% of all cancer deaths and is currently the most commoncause of cancer death among both men and women in the United States. Currently, patients are diagnosedonly when the cancer is already in a severe state because no rapid, practical, and effective screening testis available for lung cancer. In lung cancer, premalignant epithelium usually progresses to a malignanttumor through the development of dysplasia, starting from LGD and progressing to HGD, which is thentypically followed by carcinoma.152,153 Although this well-known stepwise progression could provide thebasis to survey premalignant epithelium by random biopsies, histology analyses from expert to experttypically have poor agreement, particularly for the most severe and highest-risk samples such as HGDor CIS.154 Based on such results, it appears that optical biopsies could eventually be well suited for earlydiagnosis of lung cancer and could greatly increase the survival rate of this disease.

©2003 CRC Press LLC

Page 36: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

In one study determining the potential of fluorescence spectroscopy for the diagnosis of dysplastictissues and CIS, fluorescence bronchoscopy was performed on 82 volunteers, 25 nonsmokers, 40 ex-smokers, and 17 current smokers with mean ages of 52, 55, and 49 years, respectively. Excitation of thetissue was performed using the 325-nm emission of an HeCd laser and the ratiometric analyses of theresulting fluorescence emission wavelengths were determined. Upon analysis of the results, it was foundthat the sensitivity of the autofluorescence bronchoscopy was 86%, which is 50% better than conventionalwhite-light bronchoscopy for the detection of dysplasia and CIS.93,114,

Upper Aerodigestive Tract Cancers — In order to determine the potential of laser-induced autofluores-cence for the diagnosis of dysplastic tissues in the upper aerodigestive tract, many different researchgroups have developed many types of analyses. In one study, various fluorescence excitation and emissionscans were employed for differentiation of normal mucosa from neoplastic tissues. One spectral differencewas the disappearance of an excitation peak at 330 nm in tumor tissue that existed in normal tissue,when an emission wavelength of 380 nm was used for fluorescence monitoring. Therefore, by ratioingthe excitation scan intensity at 290 nm to that of 330 nm, the resulting value could be directly correlatedto increases epithelial thickness resulting from carcinogenesis.

In addition to changes in the excitation spectrum between tumor and normal tissue, it was also foundthat the fluorescence emission intensity at 390 nm, upon excitation with 340-nm light, was also decreasedin tumor tissue relative to normal tissue. A ratio of the fluorescence emission intensity at 390 and 450nm correlated negatively with the mean epithelial thickness, again providing an indication of the stageof dysplasia.155 In a similar study, the same autofluorescence ratios were used to differentiate betweennormal and neoplastic tissues in the oral cavity and pharynx of patients with previously untreated mucosalneoplasias. Using the same ratios, researchers found that significant differences existed between theneoplastic tissue and the contralateral normal sites, leading to the thought that this technique couldrepresent a noninvasive screening method for head and neck squamous cell cancers.156

In another study, autofluorescence spectral characteristics of untreated oral and oropharyngeal lesionsin patients were studied with excitation wavelengths of 370 and 410 nm generated by a nitrogen-pumpeddye laser. Upon examining the resulting fluorescence emission of normal and neoplastic tissues, differencesin the spectral profile were noted in two regions of the spectrum for both excitation sources. However,it was found that these differences were more significant when 410-nm light is used for excitation. Byratioing the fluorescence maximum intensity of dysplastic tissue to that of normal mucosa, a quantitativemeans of differentiation has been developed for the differentiation of the two types of tissue.157

In addition to being able to determine the presence of a cancerous or precancerous lesion in a patient,it is also important to be able to determine the point or margin where the tumor ends and the healthytissue begins. Bohle and co-workers have used autofluorescence spectroscopy of tumor connective tissueto determine accurately where the tumor ends and the healthy skin begins. In this work, 365-nm lightwas used for the excitation of the tissue, and the resulting fluorescence, either from the elastic fibers of atumor or the keratinization of precancerous lesions, was used for delineation of the tissue. Contrary toprevious results in the literature, no homogeneous fluorescence gradient could be proved between darkermarginal epithelium and the brighter tumor connective fibers. The greatest differentiation between tumorand nontumor tissues was exhibited in tissues from the same patient; however, comparisons betweendifferent patients also showed promise for the margining of lesions using autofluorescence.158

Laryngeal and Oral Cancer — Like other cancers involving mucosal membranes, oral and laryngeal car-cinomas have also been studied by autofluorescence. Fluorescence spectroscopy has been used to differ-entiate normal tissue from dysplastic or cancerous tissue with a sensitivity of 90% and a specificity of88% in a training set and a sensitivity of 100% and a specificity of 98% in a validation set (Figure 28.9).159

A study of laryngeal cancer employed an HeCd laser operating at 325 nm for excitation of the sampleand an ICCD for fluorescence image collection. Images were obtained at various wavelengths with opticalfilters for wavelength discrimination and were analyzed to provide diagnostic fluorescence images of thearea of interest. This technique was used to evaluate 30 patients, of whom 18 had suspect malignanciesthat were confirmed via histopathological findings.160

©2003 CRC Press LLC

Page 37: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

Skin Diagnostics — To better understand the role of various tissue components in the autofluorescenceof skin, Gratton and co-workers161 have employed multiphoton fluorescence microscopy for the analysisof skin tissue in vivo. In this work, multiphoton fluorescence microscopy using excitation wavelengthsof 730 and 960 nm was used to image in vivo human skin cells from the surface to a depth of approximately200 µm. (Details on multiphoton fluorescence techniques are described in Chapter 11 of this handbook.)Fluorescence emission spectra and fluorescence lifetime images were obtained at selected locations nearthe surface (0 to 50 µm) and at deeper depths (100 to 150 µm) for both excitation wavelengths.

The resulting spectroscopic data suggest that reduced pyridine nucleotides and NADH are the primarysource of skin autofluorescence when using 730-nm excitation. With 960-nm excitation, a two-photon

FIGURE 28.9 In vivo detection of oral neoplasia using fluorescence spectroscopy The results demonstrate the abilityof automated data analysis algorithms to classify tissues as normal (+), dysplastic (◊), cancerous (�), or normal tissueadjacent to cancerous (∗), based upon ratios of fluorescence emission data at multiple wavelengths following excitationof the oral tissue at 350 nm. Data in (A) represent the training set for automated analyses, while (B) represents thevalidation set. The solid line at 0.28 represents the threshold for separation of normal and dysplastic or canceroustissue. Based upon these results, differentiation of normal tissue from dysplastic or cancerous tissue can be achievedwith a sensitivity of 90% and a specificity of 88% in the training set, and a sensitivity of 100% and a specificity of98% in the validation data set. (Adapted from Heintzelman, D.L. et al., Photochem. Photobiol., 72, 103, 2000.)

1

0.9

0.8

0.7

0.6

0.5

0.4

0.3

0.2

0.1

00 10 20 30 40 50 60 70

1

0.9

0.8

0.7

0.6

0.5

0.4

0.3

0.2

0.1

00 50 100 150 200 250 300

Rat

io (

472

nm/3

50 n

m)

Rat

io (

472

nm/3

50 n

m)

Sample Number

Sample Number

Cancer

DysplasiaNormal

Adjacent Normal

CancerDysplasiaNormalAdjacent Normal

A

B

©2003 CRC Press LLC

Page 38: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

fluorescence emission at 520 nm indicates the presence of a variable, position-dependent intensitycomponent caused by flavoproteins. In addition, a second fluorescence emission component that startsat 425 nm is observed when using 960-nm excitation. Such fluorescence emission at wavelengths lessthan half the excitation wavelength suggests an excitation process involving three or more photons. Thisis further confirmed by observation of a super-quadratic dependence of the fluorescence intensity on theexcitation power. Further work is still required to identify these emitting species spectroscopically;however, this study demonstrates the use of multiphoton fluorescence microscopy for functional imagingof the metabolic states of in vivo human skin cells.161

In addition to microscopic analyses, macroscopic autofluorescence-based analyses have also provideduseful information about the tissue state of skin. One property about which autofluorescence analysesof skin can provide information is the extent of photoaging that an individual’s skin has undergone.Two of the more common fluorescent components in the dermis of skin are the structural proteinselastin and collagen, which are altered by age and photoexposure. Based upon in vivo fluorescenceanalyses of skin from 28 volunteers, it was found that fluorimetry could provide a marker for the extentof photoaging that has occurred in a person. In this study, by monitoring the fluorescence emissionfrom ultraviolet irradiated skin, it was possible to determine the extent of photoaging in a way that wasindependent of the age, pigmentation, and skin thickness of the individual. Such a marker could leadto development of a technique capable of determining an individual’s risk of cancer due to ultravioletexposure from the sun.162

28.4.3.2.2 Clinical Studies and Diagnostics Using Exogenous DyesExogenous fluorophores are used for many reasons in clinical applications. The most common reason isto provide a contrasting agent that will make medical diagnoses easier, much like radionucleotides aresometimes used as contrast agents in circulation studies. However, due to the limited penetration of opticalwavelengths into biological tissues, the most common type of fluorescence analyses performed in vivo arecancer diagnoses of optically accessible tissues. For this reason the majority of clinical fluorescence diag-noses using exogenous fluorophores is in the area of cancer visualization. The most common exogenousfluorophores used for these studies are photosensitizers that have been developed for PDT treatments.

These drugs generally exhibit strong fluorescence properties, and preferentially locate in malignanttissues. Figure 28.10 illustrates the use of a fluorescence method for monitoring the production of PDTproducts produced in vivo during treatment.163 The development and applications of PDT drugs aredescribed in this handbook in Chapter 36 on mechanistic principles of photodynamic therapy, Chapter37 on synthetic strategies in designing prophyrin-based photosensitizers, and Chapter 38 on photody-namic therapy (PDT) and clinical applications.

Photosensitizers — Determining the optimal photosensitizer to use for various types of tumors is acontinuous field of investigation. In one such study, fluorescence analyses of the HPD-type photosensitizerPhotogemTM were tested in 22 patients with tumors of the lungs, larynx, and skin; gastric and esophagealcarcinoma; and cancer of the gynecological organs. Retention of the drug in the various types of tumorsand tissues was monitored by fluorescence spectroscopy after excitation of the drug with 510-nm light.The results demonstrated that tumor detection by fluorescence spectroscopy when using Photogem as acontrast agent was possible even in low selectivity of drug accumulation, which appeared to be dependenton the stage and type of the disease and the organ involved.164

Cancer Diagnostics — Liver Cancer — Monitoring and comparison of the results of ALA-induced PpIXtumor demarcation in chemically induced adenocarcinoma in the liver of rats and in an aggressive basalcell carcinoma in a patient were studied using LIF. In this study, in vivo point monitoring and fluorescencemicroscopy incorporating a CCD camera were used to study the fluorescence distribution of ALA-inducedPpIX in tumors. Fluorescence analyses were performed after i.v. injection of 30 mg/kg body weight ofALA. These analyses revealed a slightly larger concentration of PpIX in the tumor than in surroundinghealthy liver tissue or abdominal muscles in the rat. In the aggressive basal cell carcinoma of the human,a much greater concentration of PpIX was found in the visible regions of the basal cell carcinoma than

©2003 CRC Press LLC

Page 39: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

in the necrotic regions and the surrounding normal skin, thereby demonstrating selective uptake andretention of PpIX in the carcinoma.165

Oral Cancer — Diagnosis of neoplastic lesions in the oral mucosa of 11 patients was investigated usingALA-induced PpIX as a contrast agent. In this work, semiquantitative fluorescence measurements wereperformed at regular intervals of 3 h, following 15 min of continuous topical application of 0.4% ALAsolution to the lesions. Excitation of the PpIX was performed using violet light from a xenon lamp (375to 440 nm). Fluorescence images in the red region of the visible spectrum were recorded with a CCDwhile quantitative spectral analyses were performed with an OMA. From these analyses, it was foundthat PpIX accumulated earlier in the necrotic tissue than in surrounding normal tissue and that a contrastratio of 10:1 was found for tumor tissue relative to normal tissue 1 to 2 h after application of ALA.166

Esophageal Cancer — Investigation of PhotofrinTM-enhanced LIF differentiation of Barrett’s metaplasticepithelium and esophageal adenocarcinoma in five patients was performed following low-dose intravenousinjections (0.35 mg/kg body weight). In this work, LIF measurements were performed on tissue specimens

FIGURE 28.10 Fluorescence method for monitoring the production of PDT photoproducts produced in vivo duringtreatment. Immediately prior to activation of the PDT drug, a fluorescence emission spectrum is measured (A). Thenimmediately following treatment a second fluorescence spectrum is obtained (B). The fluorescent signal due to thePDT drug is designated A0 for measurements prior to irradiation, and A1 for measurements following irradiation,while C0 represents the fluorescence intensity of the photoproducts prior to irradiation and C1 the fluorescenceintensity of the photoproducts after irradiation. In order to determine the change in fluorescence intensity due tothe formation of photoproducts during PDT treatment, at the appropriate wavelength, the fluorescence intensity ofthe irradiated tissue is subtracted from the fluorescence intensity of the nonirradiated tissue, as shown in (C).(Adapted from Klinteberg, C. et al., J. Photochem. Photobiol. B, 49, 120, 1999.)

1.5

1.0

0.5

0.0550 600 650 700 750

Wavelength (nm)

Inte

nsity

(a.

u.)

1.5

1.0

0.5

0.0550 600 650 700 750

Wavelength (nm)

Inte

nsity

(a.

u.)

PDT Drug

PhotoproductsPDT Drug

Photoproducts

A B

A0C0

A1C1

0.1

0.2

0.3

0.4

0.0550 600 650 700 750

Wavelength (nm)

Inte

nsity

(a.

u.)

Differential Calculation of Photoproducts

∆P = C1 − A1C0A0

C

©2003 CRC Press LLC

Page 40: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

of normal mucosa, Barrett’s epithelium, and tumor tissue treated with Photofrin. Based upon fluorescencemeasurement ratios of the Photofrin intensity divided by the autofluorescence intensity, quantitative valuesdemonstrating the differentiation ability for the various degrees of dysplasia were determined. The meanratio ± standard deviation for each type of tissue was found to be 0.10 ± 0.058 for normal esophagealmucosa, 0.16 ± 0.073 for normal gastric mucosa, 0.205 ± 0.17 for Barrett’s epithelium with moderatedysplasia, 0.79 ± 0.54 for severe dysplasia, and 0.78 ± 0.56 for adenocarcinoma.167

Bladder Cancer — Diagnosis of bladder cancer based upon the LIF of exogenous fluorophores has alsobeen performed. In this study, a point-monitoring fluorescence diagnostic system based on a low-energypulsed laser, fiber transmission optics, and an OMA was used for the diagnosis of 24 patients with bladdermalignancies. Malignancies ranged from bladder carcinoma, CIS, and dysplasia. In order to provide abetter contrast than can be achieved using autofluorescence, the hematoporphyrin derivative, Photofrin,was injected into the tissue at a concentration of 0.35 or of 0.5 mg/kg body weight. Fluorescencemeasurements were taken 48 h after injection and a ratio of the red photosensitizer fluorescence to theblue autofluorescence of the tissue was calculated. Based upon this ratio, excellent demarcation betweenpapillary tumors and normal bladder wall tissue was achieved. In addition, these ratios allowed forobjective differentiation of certain cases of dysplasia from normal mucosa and benign exophytic lesionssuch as malakoplakia from malignant tumors.168

Skin Cancer — Due to the great success in using fluorescence spectroscopy for diagnosis of many othertypes of malignancies, LIF has also been suggested for the noninvasive diagnosis of malignant melanomas.In one study, LIF was used for real-time monitoring of the PpIX distribution in tumor tissues and in thenormal surrounding skin, before and after treatment in all patients. Based upon comparison of fluores-cence intensities from normal and tumor tissue of the same pigmentation from the same individual,PpIX distribution demonstrated excellent demarcation between tumor and normal skin of approximately15:1 for basal cell carcinoma BCC and Bowen’s disease, and 5:1 for T-cell lymphomas.169 However, thereliability of such measurements can be seriously compromised by spatial variations in the opticalproperties of the tissue that are not related to malignancy (e.g., pigmentation).

One approach to fluorescence-based analyses that minimizes this problem employs a double ratio-metric analysis procedure and the use of the photosensitizer ALA. In this technique, two types offluorescence ratios were calculated. The first of these ratios was the ratio of the skin’s fluorescence emissionbetween 660 and 750 nm to the emission between 550 and 600 nm following 405-nm excitation. Thesecond ratio was calculated by dividing the fluorescence emission between 660 and 750 nm by the emissionbetween 550 and 600 nm following excitation with 435 nm. These two excitation wavelengths were chosento be close to the fluorescence emission wavelength of ALA-induced PpIX, but some distance from theSoret excitation band of the porphyrins. Analysis of either of the two single ratios showed a significantcorrelation to the presence of a lesion and also to the color of the skin tissue. However, when the 405-nm excited ratio is divided by the 435-nm excited ratio, a value independent of skin coloration can bedetermined and can be based primarily on the presence of the ALA-induced photosensitizer. Such atechnique will enable in vivo studies of the pharmacokinetics of tumor-localizing agents in pigmentedlesions and may significantly contribute to development of a noninvasive diagnostic tool for malignantmelanoma.170

Colon Cancer — Another class of compounds beginning to be tested for in vivo tumor demarcation isfluorescently labeled antibodies. Based on previous experiments performed using nude mice, a feasibilitystudy was performed to determine whether or not LIF of fluoresceinated monoclonal antibodies againstcarcinoembryonic antigens localized specifically in human carcinoma xenografts could be used for clinicalcolon cancer diagnoses. In this study, six patients with known primary colorectal carcinoma were givenan i.v. injection of 4.5 or 9 mg of mouse–human chimeric anticarcinoembryonic antigen monoclonalantibody labeled with 0.10 to 0.28 mg of fluorescein. In addition, the monoclonal antibody was labeledwith 0.2 to 0.4 mCi of 125I. Photodetection of the tumor was performed ex vivo on surgically resectedtissues from all six patients and in vivo by fluorescence rectosigmoidoscopy for one of the six patients.

©2003 CRC Press LLC

Page 41: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

Fluorescence analyses revealed that the dye-labeled antibody localized preferentially in the tumor tissueat concentrations up to 0.059% of the injected dose per gram of tumor. This was ten times greater thanin the normal tissue, which exhibited a concentration of 0.006% of the injected dose per gram of normalmucosa. Such immunophotodiagnoses may prove very useful in the clinical setting for rapid tumordiagnoses in the colon and, potentially, other organs.171

28.5 Conclusion

This chapter illustrates the usefulness of fluorescence spectroscopy in a wide variety of medical applica-tions spanning cellular screening to tissue analysis and in vivo diagnostics. The major advantage offluorescence (as well as of other techniques such as Raman scattering, diffuse scattering, elastic scattering,and NIR absorption) is that biochemical and morphological information about the native tissue statecan be obtained without physically removing tissues. In addition, because tissue removal is not neededfor diagnostics, a more comprehensive examination of the organ can be achieved than is possible withexcised tissue. Fluorescence techniques can also be combined with imaging so that images of diseasedtissue sites vs. normal sites can be constructed. Thus, these diagnostic techniques inherently providesuperior coverage and do not suffer from the sampling errors that often occur with biopsy or cytology.Real-time diagnostic information is a significant added benefit.

At the early stage of development, fluorescence diagnostic techniques need to be verified by comparisonwith pathology, the current “gold standard,” which also varies in reliability. Thus, the sensitivity andspecificity of spectroscopic diagnostics depends upon the reliability of pathology in each specific desiredapplication. If “optical biopsy” is to have a role in clinical diagnosis, the problem becomes which goldstandard to use for calibrating optical measurements. Using pathology as the gold standard for intraep-ithelial neoplasia (preinvasive cancer) has its own difficulties because pathologists often disagree on theirinterpretations of these lesions, with the level of consistency varying for different organs. Therefore,building decision-making boundaries using optical measurements based on conventional pathology isproblematic. Because under- and over-diagnosis are undesirable, it is imperative to establish objectiveand standardized pathological classification systems for grading preinvasive lesions for each separateorgan area where these new optical technologies may be applied.

Another important application for optical diagnostic technologies such as fluorescence is the possibleuse of these techniques for guidance of surgical intervention and treatment. In such surgical-assistapplications, the ability of fluorescence diagnostic technologies to provide real-time information wouldbe critically useful. Fluorescence techniques may also be used to provide real-time assessment of tissueresponse to therapy — as in the assessment of tissue viability and necrosis in thermal, laser interstitialtherapy, or PDT.

Fluorescence technologies could also be used for noninvasive measurement of concentrations ofvarious drugs and biological species in tissues. This capability would provide a variety of benefits inmedical research. One example of such a benefit is the use of chemotherapy drugs for treatment of variouscancers. The therapeutic benefit is determined by the concentration of the drug in the tissues of thetargeted organ or site; currently, the only minimally invasive check available to the oncologist is to trackthe blood serum concentration and to assume a relationship between the amount of drugs at the targetorgans and the tissue concentration. More generally, the ability to track concentrations of compoundsin tissue noninvasively would be a tremendous advantage. Optical methods such as fluorescence couldbypass many tedious and time-consuming trials that attempt to relate dosage to metabolic rates andtarget organ concentrations. Because of all of these desirable features, it is evident that fluorescencespectroscopy can be a powerful tool for the medical researcher to obtain a better understanding of thedisease process and for the physician to perform real-time in vivo diagnoses and, ultimately, providetreatment at the point of care.

©2003 CRC Press LLC

Page 42: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

Acknowledgments

This work was sponsored by the National Institutes of Health (RO1 CA88787-01) and by the U.S.Department of Energy (DOE) Office of Biological and Environmental Research, under contract DEAC05-00OR22725 with UT-Battelle, LLC. T.V.D. acknowledges contributions from Bergein F. Overholt andMasoud Panjehpour at the Thomson Cancer Survival Center.

References

1. Vo-Dinh, T., Room Temperature Phosphorimetry for Chemical Analysis, John Wiley & Sons, NewYork, 1984.

2. McGlynn, S.P., Azumi, T., and Kinoshita, M., Molecular Spectroscopy of the Triplet State, Prentice-Hall, Engelwood Cliffs, NJ, 1969.

3. Vo-Dinh, T., Multicomponent analysis by synchronous luminescence spectrometry, Anal. Chem.,50, 396, 1978.

4. Vo-Dinh, T., Synchronous luminescence spectroscopy — methodology and applicability, Appl.Spectrosc., 36, 576, 1982.

5. Wolfbeis, O.S., Fluorescence or organic natural products, in Molecular Luminescence Spectroscopy,Schulman, S.G.J., Ed., John Wiley & Sons, New York, 1993, p. 167.

6. Richards-Kortum, R. and Sevick-Muraca, E., Quantitative optical spectroscopy for tissue diagnosis,Annu. Rev. Phys. Chem., 47, 555, 1996.

7. Bottiroli, G.C., Locatelli, A.C.., Marchesini, R., Pignoli, E., Tomatis, S., Cuzzoni, C., Dipalma, S.,Dalfante, M., and Spinelli, P, Natural fluorescence of normal and neoplastic human colon: acomprehensive ex vivo study, Lasers Surg. Med., 16, 48, 1995.

8. Fujimoto, D., Akiba, K.Y., and Nakamura, N., Isolation and characterization of a fluorescentmaterial in bovine Achilles-tendon collagen, Biochem. Biophys. Res. Commun., 76, 1124, 1977.

9. Wilson, B., Laser reflectance spectroscopy of tissue, in Optronic Techniques in Diagnostic andTherapeutic Medicine, Pratesi, R., Ed., Plenum Press, New York, 1991.

10. Wagnières, G.A., Star, W.M., and Wilson, B.C., In vivo fluorescence spectroscopy and imaging foroncological applications, Photochem. Photobiol., 68, 603, 1998.

11. Lakowicz, J.R., Principles of Fluorescence Spectroscopy, Plenum Press, New York, 1985.12. Masters, B.R. and Chance, B., in Fluorescent and Luminescent Probes for Biological Activity, Maon,

W.T., Ed., Academic Press, London, 1993, p. 44.13. Tsuchida, M., Miura, T., and Aibara, K., Lipofuscin and lipofuscin-like substances, Chem. Phys.

Lipids, 44, 297, 1987.14. Eldred, G.E., Miller, G.V., Stark, W.S., and Feeney-Burns, L., Lipofuscin — resolution of discrepant

fluorescence data, Science, 216, 757, 1982.15. Millar, D.P., Time-resolved fluorescence spectroscopy, Curr. Opin. Struct. Biol., 6, 637, 1996.16. Viallet, P.M., Vo-Dinh, V., Bunde, T., Ribou, A.C., Vigo, J., and Salmon, J.M., Fluorescent molecular

reporter for the 3-D conformation of protein subdomains: the Mag-Indo system, J. Fluorescence,9, 153, 1999.

17. Viallet, P.M., Vo-Dinh, T., Ribou, A.C., Vigo, J., and Salmon, J.M., Native fluorescence and Mag-Indo-1-protein interaction as tools for probing unfolding and refolding sequences of the bovineserum albumin subdomain in the presence of guanidine hydrochloride, J. Protein Chem., 19, 431,2000.

18. Tyagi, S. and Kramer, F.R., Molecular beacons: probes that fluoresce upon hybridization, Nat.Biotechnol., 14, 303, 1996.

19. Gao, W.W., Tyagi, S., Kramer, F.R., and Goldman, E., Messenger RNA release from ribosomesduring 5′-translational blockage by consecutive low-usage arginine but not leucine codons inEscherichia coli, Mol. Microbiol., 25, 707, 1997.

©2003 CRC Press LLC

Page 43: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

20. Tyagi, S., Bratu, D.P., and Kramer, F.R., Multicolor molecular beacons for allele discrimination,Nat. Biotechnol., 16, 49, 1998.

21. Tyagi, S., Marras, S.A.E., and Kramer, F.R., Wavelength-shifting molecular beacons, Nat. Biotech-nol., 18, 1191, 2000.

22. Protio, A.E. and Sarniak, J., Fluorescence of HpD for tumor detection and dosimetery in photo-radiation therapy, in Porphyrin Localization and Treatment of Tumors, Doiron, D.R. and Gomer,C.J., Eds., Alan R. Liss, New York, 1984, p. 163.

23. Lam, S.H.J. and Palcic, B., Detection of lung cancer by ratio fluorimetry with and without photofrinII, Proc. SPIE, 1201, 561, 1990.

24. Tassetti, V.H., Sowinska, M., Evrard, S., Heisel, F., Cheng, L.Q., Miehe, J.A., Marescaux, J., andAprahamian, M., In vivo laser-induced fluorescence imaging of a rat pancreatic cancer withpheophorbide-a, Photochem. Photobiol., 65, 997, 1997.

25. Kim, R.Y.H., Flotte, T.J., Gragoudas, E.S., and Young, L.H.Y., Digital angiography of experimentalchoroidal melanomas using benzoporphyrin derivative, Am. J. Ophthalmol., 123, 810 , 1997.

26. Kollias, N.L.H., Wimberly, J., and Anderson, R.R., Monitoring of benzoporphyrin derivativemonoacid ring a (bpd-ma) in skin tumors by fluorescence during photodynamic therapy, Proc.SPIE, 1881, 41, 1993.

27. Peyman, G.A., Moshfeghi, D.M., Moshfeghi, A., Khoobehi, B., Doiron, D.R., Primbs, G.B., andCrean, D.H., Photodynamic therapy for choriocapillaris using tin ethyl etiopurprin (SnET2), Oph-thalmic Surg. Lasers, 28, 409, 1997.

28. Koren, H., Schenk, G.M., Jindra, R.H., Alth, G., Ebermann, R., Kubin, A., Koderhold, G., andKreitner, M., Hypericin in phototherapy, J. Photochem. Photobiol., B36, 113, 1996.

29. Diwu, Z., Novel therapeutic and diagnostic applications of hypocrellins and hypericins, Photochem.Photobiol., 61, 529, 1995.

30. Dets, S.M, Buryi, A.N., Melnik, I.S., Joffe, A.Y., and Rusina, T.V., Laser-induced fluorescencedetection of stomach cancer using hypericin, Proc. SPIE, 2926, 51, 1996.

31. Peng, Q., Berg, K., Moan, J., Kongshaug, M., and Nesland, J.M., 5-Aminolevulinic acid-based photo-dynamic therapy — principles and experimental research, Photochem. Photobiol., 65, 235, 1997.

32. Battle, A.M.D.C., Phorophyrins, phorphyrias, cancer and photodynamic therapy: a model of car-cinogenesis, J. Photochem. Photobiol., B20, 5, 1993.

33. Cox, G.S., Bobillier, C., and Whitten, D.G., Photooxidation and singlet oxygen sensitization byphotophyrin ix and its photoxidiation products, Photochem. Photobiol., 36, 401, 1982.

34. Peng, Q., Warloe, T., Berg, K., Moan, J., Kongshaug, M., Giercksky, K.E., and Nesland, J.M., 5-Aminolevunic acid based photodynamic therapy: clinical research and future challenges, Cancer,79, 2282, 1997.

35. Marcus, S.L., Solbel, R.S., Golub, A.L., Carroll, R.L., Lundahl, S., and Shulman, D.G., Photodynamictherapy (PDT) and photodiagnosis (PD) using endogenous photosensitization induced by 5-aminolevulinic acid (ALA): current clinical and developmental status, Laser Med. Surg., 14, 59,1996.

36. Schwartz, S., Historical perspectives, in Photodynamic Therapy: Basic Priniciples and Clinical Appli-cations, Henderson, B.W. and Dougherty, T.J., Eds., Marcel Dekker, New York, 1992, p. 1.

37. Sacks, P.G., Savage, H.E., Levine, J., Kolli, V.R., Alfano, R.R., and Schantz, S.P., Native cellularfluorescence identifies terminal squamous differentiation of normal oral epithelial cells in culture:a potential chemoprevention biomarker, Cancer Lett., 104, 171, 1996.

38. Zhang, J.C., Savage, H.E., Sacks, P.G., Delohery, T., Alfano, R.R., Katz, A., and Schantz, S.P., Innatecellular fluorescence reflects alterations in cellular proliferation, Lasers Surg. Med., 20, 319, 1997.

39. Anidjar, M., Cussenot, O., Blais, J., Bourdon, O., Avrillier, S., Ettori, D., Villette, J.M., Fiet, J., Teillac,P., and LeDuc, A., Argon laser induced autofluorescence may distinguish between normal andtumor human urothelial cells: a microspectrofluorimetric study., J. Urol., 155, 1771, 1996.

©2003 CRC Press LLC

Page 44: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

40. Harth, R., Gerlach, M., Riederer, P., and Gotz, M.E., A highly sensitive method for the determina-tion of protein bound 3,4-dihydroxyphenylalanine as a marker for post-translational proteinhydroxylation in human tissues ex vivo, Free Radical Res., 35, 167, 2001.

41. Alfano, R.R., Tata, D.B., Cordero, J., Tomashefsky, P., Longo, F.W., and Alfano, M.A., Laser-inducedfluorescence spectroscopy from native cancerous and normal tissue, IEEE J. Quantum Electron.,20, 1507, 1984.

42. Uziel, M., Ward, R.J., and Vo-Dinh, T., Synchronous fluorescence measurement of bap metabolitesin human and animal urine, Anal. Lett., 20, 761, 1987.

43. Watts, W.E., Isola, N.R., Frazier, D., and Vo-Dinh, T., Differentiation of normal and neoplastic cellsby synchronous fluorescence: rat liver epithelial and rat hepatoma cell models, Anal. Lett., 32, 2583,1999.

44. Amano, T., Kunimi, K., and Ohkawa, M., Fluorescence spectra from human semen and theirrelationship with sperm parameters, Arch. Androl., 36, 9, 1996.

45. Xu, H., Aylott, J., and Kopelman, R., Sol-gel PEBBLE sensors for biochemical analysis inside livingcells, Abstr. Papers Am. Chem. Soc., 219, 97-ANYL, 2000.

46. Tan, W.H., Thorsrud, B.A., Harris, C., and Kopelman, R., Real time pH measurements in the intactrat conceptus using ultramicrofiber-optic sensors, in Polymers in Sensors, American ChemicalSociety, Washington, D.C., 1998, p. 266.

47. Clark, H.A., Hoyer, M., Philbert, M.A., and Kopelman, R., Optical nanosensors for chemicalanalysis inside single living cells. 1. Fabrication, characterization, and methods for intracellulardelivery of PEBBLE sensors, Anal. Chem., 71, 4831, 1999.

48. Clark, H.A., Kopelman, R., Tjalkens, R., and Philbert, M.A., Optical nanosensors for chemicalanalysis inside single living cells. 2. Sensors for pH and calcium and the intracellular applicationof PEBBLE sensors, Anal. Chem., 71, 4837, 1999.

49. Tan, W.H., Kopelman, R., Barker, S.L.R., and Miller, M.T., Ultrasmall optical sensors for cellularmeasurements, Anal. Chem., 71, 606A, 1999.

50. Cullum, B.M., Griffin, G.D., Miller, G.H., and Vo-Dinh, T., Intracellular measurements in mam-mary carcinoma cells using fiber-optic nanosensors, Anal. Biochem., 277, 25, 2000.

51. Cullum, B.M. and Vo-Dinh, T., The development of optical nanosensors for biological measure-ments, Trends Biotechnol., 18, 388, 2000.

52. Vo-Dinh, T., Alarie, J.P., Cullum, B.M., and Griffin, G.D., Antibody-based nanoprobe for measure-ment of a fluorescent analyte in a single cell, Nat. Biotechnol., 18, 764, 2000.

53. Kong, J.A.R.,D.P., Quantitative in situ image analysis of apoptosis in well and poorly differentiatedtumors from rat liver, Am. J. Pathol., 147, 1626, 1995.

54. Balazs, M.S.J., Lee, W.C., Haugland, R.P., Guzikowski, A.P., Fulwyler, M.J., Damjanovich, S., Feurst-ein, B.G., and Pershadsingh, H.A., Fluorescent tetradecanoylphorbol acetate: a novel probe ofphorbol ester binding domains, J. Cell Biochem., 46, 266, 1991.

55. Andeoni, A., Bottiroli, G., Colasanti, A., Giangare, M.C., Riccio, P., Roberti, G. and Vaghi, P.,Fluorochromes with long-lived fluorescence as potential labels for pulsed laser immunocytofluo-rometry: photophysical characterization of pyrene derivatives, J. Biochem. Biophys. Methods, 29,157, 1994.

56. Fritsch, C., Batz, J., Bolsen, K., Schulte, K.W., Zumdick, M., Ruzicka, T., and Goerz, G., Ex vivoapplication of delta-aminolevulinic acid induces high and specific porphyrin levels in human skintumors: possible basis for selective photodynamic therapy, Photochem. Photobiol., 66, 114, 1997.

57. DaCosta, R.S.L., Andersson, H.M.S., Cirocco, M., Kandel, G., Kortan, P., Haber, G., Marcon, N.E.,and Wilson, B.C., Correlation of autofluorescence (AF) and ultrastructures of ex vivo colorectaltissues and isolated living epithelial cells from primary cell cultures (PCC) of normal colon andhyperplastic and dysplastic polyps: implications for early diagnosis, altered cell metabolism andcytopathology, Gastroenterology, 116, G1721, 1999.

©2003 CRC Press LLC

Page 45: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

58. Han, I., Saito, H., Fukatsu, K., Inoue, T., Yasuhara, H., Furukawa, S., Matsuda, T., Lin, M.T., andIkeda, S., Ex vivo fluorescence microscopy provides simple and accurate assessment of neutrophil-endothelial adhesion in the rat lung, Shock, 16, 143, 2001.

59. Sculean, A., Auschill, T.M., Donos, N., Brecx, M., and Arweiler, N.B., Effect of an enamel matrixprotein (Emdogain (R)) on ex vivo dental plaque vitality, J. Clin. Periodontol., 28, 1074, 2001.

60. Alfano, R.R., Lam, W., Zarrabi, H.J., Alfano, M.A., Cordero, J., Tata, D.B., and Swenberg, C.E.,Human teeth with and without caries studied by laser scattering, fluorescence, and absorption-spectroscopy, IEEE J. Quantum Electron., 20, 1512, 1984.

61. Alfano, R.R., Tang, G.C., Pradhan, A., Lam, W., Choy, D.S.J., and Opher, E., Fluorescence spectrafrom cancerous and normal human-breast and lung tissues, IEEE J. Quantum Electron., 23, 1806,1987.

62. Tang, G.C., Pradhan, A., Sha, W., Chen, J., Liu, C.H., Wahl, S.J., and Alfano, R.R., Pulsed and cwlaser fluorescence spectra from cancerous, normal, and chemically treated normal human-breastand lung tissues, Appl. Opt., 28, 2337, 1989.

63. Ingrams, D.R., Dhingra, J.K., Roy, K., Perrault, D.F., Bottrill, I.D., Kabani, S., Rebeiz, E.E., Pankra-tov, M.M., Shapshay, S.M., Manoharan, R., Itzkan, I., and Feld, M.S., Autofluorescence character-istics of oral mucosa, Head Neck J. Sci. Specialties Head Neck, 19, 27, 1997.

64. Vo-Dinh, T., Panjehpour, M., Overholt, B.F., and Buckley, P., Laser-induced differential fluorescencefor cancer diagnosis without biopsy, Appl. Spectrosc., 51, 58, 1997.

65. Vo-Dinh, T., Panjehpour, M., and Overholt, B.F., Laser-induced fluorescence for esophageal cancerand dysplasia diagnosis, in Advances in Optical Biopsy and Optical Mammography, New YorkAcademy of Sciences, New York, 1998, p. 116.

66. Richards-Kortum, R., Mitchell, M.F., Ramanujam, N., Mahadevan, A., and Thomsen, S., In vivofluorescence spectroscopy: potential for non-invasive, automated diagnosis of cervical intraepithe-lial neoplasia and use as a surrogate endpoint biomarker, J. Cell Biochem., Suppl. 19, 111, 1994.

67. Lohmann, W., Mussman, J., Lohmann, C., and Kunzel, W., Native fluorescence of cervix uteri asa marker for dysplasia and invasive carcinoma, Eur. J. Obstet. Gynecol. Reprod. Biol., 31, 249, 1989.

68. Romer, T.J., Fitzmaurice, M., Cothren, R.M., Richardskortum, R., Petras, R., Sivak, M.V., andKramer, J.R., Laser-induced fluorescence microscopy of normal colon and dysplasia in colonicadenomas — implications for spectroscopic diagnosis, Am. J. Gastroenterol., 90, 81, 1995.

69. Fiarman, G.S., Nathanson, M.H., West, A.B., Deckelbaum, L.I., Kelly, L., and Kapadia, C.R., Dif-ferences in laser-induced autofluorescence between adenomatous and hyperplastic polyps andnormal colonic mucosa by confocal microscopy, Dig. Dis. Sci., 40, 1261, 1995.

70. Hegedus, Z.L. and Nayak, U., Relative fluorescence intensities of human plasma soluble melaninsin normal adults, Arch. Int. Physiol. Biochim. Biophys., 102, 311, 1994.

71. Marchesini, R., Brambilla, M., Pignoli, E., Bottiroli, G., Croce, A.C., Dal Fante, M., Spinelli, P.,and diPalma, S., Light-induced fluorescence spectroscopy of adenomas, adenocarcinomas and non-neoplastic mucosa in human colon I. In vitro measurements, J. Photochem. Photobiol. B., 14, 219,1992.

72. Richards-Kortum, R., Rava, R.P., Petras, R.E., Fitzmaurice, M., Sivak, M., and Feld, M.S., Spectro-scopic diagnosis of colonic dysplasia, Photochem. Photobiol., 53, 777, 1991.

73. Kapadia, C.R., Cutruzzola, F.W., O’Brien, K.M., Stetz, M.L., Enriquez, R., and Deckelbaum, L.I.,Laser-induced fluoresence spectroscopy of human colonic mucosa, Gastroenterology, 99, 150, 1990.

74. Chwirot, B.W., Chwirot, S., Jedrzejczyk, W., Jackowski, M., Raczynska, A.M., Winczakiewicz, J.,and Dobber, J., Ultraviolet laser-induced fluorescence of human stomach tissues: detection ofcancer tissues by imaging techniques, Lasers Surg. Med., 21, 149, 1997.

75. Laufer, G.W.G., Hohla, K., Horvat, R., Henke, K.H., Buchelt, M., Wutzl, G., and Wolner, E., Excimerlaser-induced simultaneous ablation and spectral identification of normal and atheroscleroticarterial tissue layers, Circulation, 78, 1031, 1988.

©2003 CRC Press LLC

Page 46: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

76. Baraga, J.J., Rava, R.P., Taroni, P., Kittrell, C., Fitzmaurice, M., and Feld, M.S., Laser inducedfluorescence spectroscopy of normal and atherosclerotic human aorta using 306 to 310 nm exci-tation, Lasers Surg. Med., 10, 245, 1990.

77. Bosshart, F.U., Hess, O.M., Wyser, J., Mueller, A., Schneider, J., Niederer, P., Anliker, M., andKrayenbuehl, H.P., Fluorescence spectroscopy for identification of atherosclerotic tissue, Cardio-vasc. Res., 26, 620, 1992.

78. Lucas, A., Radosavljevic, M.J., Lu, E., and Gaffney, E.J., Characterization of human coronary arteryatherosclerotic plaque fluorescence emission, Can. J. Cardiol., 6, 219, 1990.

79. Richards-Kortum, R., Rava, R.P., Fitzmaurice, M., Kramer, J.R., and Feld, M.S., 476 nm excitedlaser-induced fluorescence spectroscopy of human coronary arteries applications in cardiology,Am. Heart J., 122, 1141, 1991.

80. Verbunt, R., Fitzmaurice, M.A., Kramer, J.R., Ratliff, N.B., Kittrell, C., Taroni, P., Cothren, R.M.,Baraga, J., and Feld, M., Characterization of ultraviolet laser-induced autofluorescence of ceroiddeposits and other structures in atherosclerotic plaques as a potential diagnostic for laser angio-surgery, Am. Heart J., 123, 208, 1992.

81. Fitzmaurice, M., Bordagary, J.O., Engelmann, G.L., Richards-Kortum, R., Kolubayev, T., Feld, M.S.,Ratliff, N.B., and Kramer, J.R., Argon ion laser-excited autofluorescence in normal and atheroscle-rotic aorta and coronary arteries: morphologic studies, Am. Heart J., 118, 1028, 1989.

82. Chaudhry, H.W., Richards-Kortum, R., Kolubayev, T., Kittrell, C., Partovi, F., Kramer, J.R., andFeld, M.S., Alteration of spectral characteristics of human artery wall caused by 476-nm laserirradiation, Lasers Surg. Med., 9, 572, 1989.

83. Andersson-Engels, S.J.J., Svanberg, K., and Svanberg, S., Fluorescence imaging and point measure-ments of tissue: applications to the demarcation of malignant tumors and atherosclerotic lesionsfrom normal tissue, Photochem. Photobiol., 53, 807, 1991.

84. Andersson-Engels, S.J.J., Stenram, U., Svanberg, K., and Svanberg, S., Time-resolved laser-inducedfluorescence spectroscopy for enhanced demarction of human atherosclerotic plaques, J. Photo-chem. Photobiol. B., 4, 363, 1990.

85. Perk, M., Flynn, G.J., Gulamhusein, S., Wen, Y., Smith, C., Bathgate, B., Tulip, J., Parfrey, N.A.,and Lucas, A., Laser induced fluorescence identification of sinoatrial and atrioventricular nodalconduction tissue, Pacing Clin. Electrophysiol., 16, 1701, 1993.

86. van der Veen, M.H. and ten Bosch, J.J., Autofluorescence of bulk sound and in vitro demineralizedhuman root dentin, Eur. J. Oral Sci., 103, 375, 1995.

87. Samoszuk, M., Eosinophils and human cancer, Histol. Histopathol., 12, 807, 1997.88. Barnes, D.A.S., Thomsen, S., Fitzmaurice, M., and Richards-Kortum, R., A characterization of the

fluorescent properties of circulating human eosinophils, Photochem. Photobiol., 58, 297, 1993.89. Pompella, A. and Comporti, M., Imaging of oxidative stress at subcellular level by confocal laser

scanning microscopy after fluorescent derivitization of cellular carbonyls, Am. J. Pathol., 142, 1353,1993.

90. Richards-Kortum, R., Mehta, A., Hayes, G., Cothren, R., Kolubayev, T., Kittrell, C., Ratliff, N.B.,Kramer, J.R., and Feld, M.S., Spectral diagnosis of atherosclerosis using an optical fiber lasercatheter, Am. Heart J., 118, 381, 1989.

91. Alfano, R.R. and Yao, S.S., Human teeth with and without dental caries studied by visible lumi-nescent spectroscopy, J. Dent. Res., 60, 120, 1981.

92. Zellweger, M., Goujon, D., Conde, R., Forrer, M., van den Bergh, H., and Wagnières, G., Absoluteautofluorescence spectra of human healthy, metaplastic, and early cancerous bronchial tissue invivo, Appl. Opt., 40, 3784, 2001.

93. Zellweger, M., Grosjean, P., Goujon, D., Monnier, P., van den Bergh, H., and Wagnières, G., In vivoautofluorescence spectroscopy of human bronchial tissue to optimize the detection and imagingof early cancers, J. Biomed. Opt., 6, 41, 2001.

©2003 CRC Press LLC

Page 47: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

94. Wang, T.D., Crawford, J.M., Feld, M.S., Wang, Y., Itzkan, I., and Van Dam, J., In vivo identificationof colonic dysplasia using fluorescence endoscopic imaging, Gastrointest. Endosc., 49, 447, 1999.

95. Wennberg, A.M., Gudmundson, F., Stenquist, B., Ternesten, A., Molne, L., Rosen, A., and Larko,O., In vivo detection of basal cell carcinoma using imaging spectroscopy, Acta Dermato-Venereolog.,79, 54, 1999.

96. Schantz, S.P., Kolli, V., Savage, H.E., Yu, G.P., Shah, J.P., Harris, D.E., Katz, A., Alfano, R.R., andHuvos, A.G., In vivo native cellular fluorescence and histological characteristics of head and neckcancer, Clin. Cancer Res., 4, 1177, 1998.

97. Major, A.L., Rose, G.S., Chapman, C.F., Hiserodt, J.C., Tromberg, B.J., Krasieva, T.B., Tadir, Y.,Haller, U., DiSaia, P.J., and Berns, M.W., In vivo fluorescence detection of ovarian cancer in theNuTu-19 epithelial ovarian cancer animal model using 5-aminolevulinic acid (ALA), Gynecol.Oncol., 66, 122, 1997.

98. Andersson-Engels, S., Afklinteberg, C., Svanberg, K., and Svanberg, S., In vivo fluorescence imagingfor tissue diagnostics, Phys. Med. Biol., 42, 815, 1997.

99. Ramanujam, N., Chen, J.X., Gossage, K., Richards-Kortum, R., and Chance, B., Fast and nonin-vasive fluorescence imaging of biological tissues in vivo using a flying-spot scanner, IEEE Trans.Biomed. Eng., 48, 10341, 2001.

100. Brancaleon, L., Durkin, A.J., Tu, J.H., Menaker, G., Fallon, J.D., and Kollias, N., In vivo fluorescencespectroscopy of nonmelanoma skin cancer, Photochem. Photobiol., 73, 178, 2001.

101. Koenig, F., Knittel, J., and Stepp, H., Diagnosing cancer in vivo, Science, 292, 1401, 2001.102. Brewer, M., Utzinger, U., Silva, E., Gershenson, D., Bast, R.C., Follen, M., and Richards-Kortum,

R., Fluorescence spectroscopy for in vivo characterization of ovarian tissue, Lasers Surg. Med., 29,128, 2001.

103. Panjehpour, M., Overholt, B.F., Vo-Dinh, T., Farris, C., and Sneed, R., Fluorescence spectroscopyfor detection of malignant-tissue in the esophagus, Gastroenterology, 104, A439, 1993.

104. Panjehpour, M., Overholt, B.F., Schmidhammer, J.L., Farris, C., Buckley, P.F., and Vo-Dinh, T.,Spectroscopic diagnosis of esophageal cancer — new classification model, improved measurementsystem, Gastrointest. Endosc., 41, 577, 1995.

105. Panjehpour, M., Overholt, B.F., Vo-Dinh, T., Haggitt, R.C., Edwards, D.H., and Buckley, F.P.,Endoscopic fluorescence detection of high-grade dysplasia in Barrett’s esophagus, Gastroenterology,111, 93, 1996.

106. Panjehpour, M., Overholt, B.F., Vo-Dinh, T., Haggitt, R.C., Edwards, D.H., Buckley, F.P., andDecosta, J.F., Fluorescence spectroscopy for detection of dysplasia in Barrett’s esophagus, Gastro-enterology, 110, A574, 1996.

107. Overholt, B.F., Panjehpour, M., Vo-Dinh, T., Farris, C., Schmidhammer, J.L., Sneed, R.E., andBuckley, P.F., Spectroscopic diagnosis of esophageal cancer — improved technique, Gastroenterol-ogy, 106, A425, 1994.

108. Richards-Kortum, R. and Sevick-Muraca, E., Quantitative optical spectroscopy for tissue diagnosis,Annu. Rev. Phys. Chem., 47, 555, 1996.

109. Ramanujam, N., Fluorescence spectroscopy of neoplastic and non-neoplastic tissues, Neoplasia, 2,89, 2000.

110. Ramanujam, N., Mitchell, M.F., Mahadevan-Jansen, A., Thomsen, S.L., Staerkel, G., Malpica, A.,Wright, T., Atkinson, N., and Richards-Kortum, R., Cervical precancer detection using a multi-variate statistical algorithm based on laser-induced fluorescence spectra at multiple excitationwavelengths, Photochem. Photobiol., 64, 720, 1996.

111. Lam, S. and Shibuya, H., Early diagnosis of lung cancer, Clin. Chest Med., 20, 53, 1999.112. Lam, S. and Palcic, B., Autofluorescence bronchoscopy in the detection of squamous metaplasia

and dysplasia in current and former smokers, J. Natl. Cancer Inst., 91, 561, 1999.113. Lam, S., Macaulay, C., Hung, J., Leriche, J., Profio, A.E., and Palcic, B., Detection of dysplasia and

carcinoma in situ with a lung imaging fluorescence endoscope device, J. Thorac. Cardiovasc. Surg.,105, 1035, 1993.

©2003 CRC Press LLC

Page 48: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

114. Lam, S., Hung, J.Y.C., Kennedy, S.M., Leriche, J.C., Vedal, S., Nelems, B., Macaulay, C.E., and Palcic,B., Detection of dysplasia and carcinoma in situ by ratio fluorometry, Am. Rev. Respir. Dis., 146,1458, 1992.

115. Alfano, R.R. and Alfano, M.A., Medical diagnostics — a new optical frontier, Photonics Spectra,19, 55, 1985.

116. Alfano, R.R., Tang, G.C., Pradhan, A., and Wenling, S., Investigation of optical spectroscopy ofcancerous and normal human tissues, J. Electrochem. Soc., 135, C387, 1988.

117. Alfano, R.R., Tomaselli, V.P., Beuthan, J., Feld, M.S., Flotte, T.J., Fujimoto, J.G., and Thomsen, S.,Advances in optical biopsy and optical mammography — panel discussion — review and summaryof presentations, in Advances in Optical Biopsy and Optical Mammography, New York Academy ofSciences, New York, 1998, p. 194.

118. Alfano, R.R., Advances in optical biopsy and optical mammography — closing remarks, in Advancesin Optical Biopsy and Optical Mammography, New York Academy of Sciences, New York, 1998,p. 197.

119. Vo-Dinh, T., Panjehpour, M., Overholt, B.F., Farris, C., Buckley, F.P., and Sneed, R., In-vivo cancer-diagnosis of the esophagus using differential normalized fluorescence (DNF) indexes, Lasers Surg.Med., 16, 41, 1995.

120. Vo-Dinh, T., Panjehpour, M., Overholt, B.F., and Buckley, P., Laser-induced differential fluorescencefor cancer diagnosis without biopsy, Appl. Spectrosc., 51, 58, 1997.

121. Braichotte, D., Savary, J.F., Glanzmann, T., Westermann, P., Folli, S., Wagnieres, G., Monnier, P.,and Vandenbergh, H., Clinical pharmacokinetic studies of tetra(meta-hydroxyphenyl)chlorin insquamous-cell carcinoma by fluorescence spectroscopy at 2 wavelengths, Int. J. Cancer, 63, 198,1995.

122. Braichotte, D.R., Wagnières, G.A., Bays, R., Monnier, P., and Vandenbergh, H.E., Clinical pharma-cokinetic studies of photofrin by fluorescence spectroscopy in the oral cavity, the esophagus, andthe bronchi, Cancer, 75, 2768, 1995.

123. Andrejevic, S., Savary, J.-F., Fontolliet, C., Monnier, P., and van den Bergh, H.E., 7,12-Dimethyl-benz(a)anthracene-induced early squamous-cell carcinoma in the golden Syrian hamster: evalua-tion of an animal model and comparison with early forms of human squamous-cell carcinoma inthe upper aerodigestive tract., Int. J. Exp. Pathol., 77, 7, 1996.

124. Schantz, S.P. and Alfano, R.R., Tissue autofluorescence as an intermediate end-point in cancerchemoprevention trials, J. Cell. Biochem., 64, 199, 1993.

125. Glasgold, R., Glasgold, M., Savage, H., Pinto, J., Alfano, R., and Schantz, S., Tissue autofluorescenceas an intermediate end-point in NMBA-induced esophageal carcinogenesis, Cancer Lett., 82, 33,1994.

126. Chung, Y.G., Schwartz, J.A., Gardner, C.M., Sawaya, R.E. and Jacques, S.L., Diagnostic potentialof laser-induced autofluorescence emission in brain tissue, J. Korean Med. Sci., 12, 135, 1997.

127. Oraevsky, A.A., Jacques, S.L., Pettit, G.H., Sauerbrey, R.A., Tittel, F.K., Nguy, J.H., and Henry, P.D.,XeCl laser-induced fluorescence of atherosclerotic arteries. Spectral similarities between lipid-richlesions and peroxidized lipoproteins, Circ. Res., 72, 84, 1993.

128. van Leengoed, E.V.J., van der Veen, N., van der Berg-Blok, A., Marijnissen, H., and Star, W., Tissue-localizing properties of some photosensitizers studied by in vivo fluorescence imaging, J. Photochem.Photobiol., B. 6, 111, 1990.

129. Johansson, J., Berg, R., Svanberg, K., and Svanberg, S., Laser-induced fluorescence studies of normaland malignant tumour tissue of rat following intravenous injection of delta-amino levulinic acid,Lasers Surg. Med., 20, 272, 1997.

130. Svanberg, K., Kjellen, E., Ankerst, J., Montan, S., Sjoholm, E., and Svanberg, S., Fluorescence studiesof hematoporphyrin derivative in normal and malignant rat-tissue, Cancer Res., 46, 3803, 1986.

131. Andersson-Engels, S., Ankerst, J., Johansson, J., Svanberg, K., and Svanberg, S., Laser-inducedfluorescence in malignant and normal tissue of rats injected with benzoporphyrin derivative,Photochem. Photobiol., 57, 978, 1993.

©2003 CRC Press LLC

Page 49: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

132. Nilsson, H.J.J., Svanberg, K., Svanberg, S., Jori, G., Reddi, E., Segalla, A., Gust, D., Moore, A.L.,and Moore, T.A., Laser-induced fluorescence studies of the biodistribution of carotenoporphyrinsin mice, Br. J. Cancer, 76, 355, 1997.

133. Pathak, I., Davis, N.L., Hsiang, Y.N., Quenville, N.F., and Palcic, B., Detection of squamous neo-plasia by fluorescence imaging comparing porfimer sodium fluorescence to tissue autofluorescencein the hamster cheek pouch model, Am. J. Surg., 170, 423, 1995.

134. Tsai, J.C., Kao, M.C., and Hsiao, Y.Y., Fluorospectral study of the rat brain and glioma in vivo,Lasers Surg. Med., 13, 321, 1993.

135. Svanberg, K., Liu, D.L., Wang, I., Andersson-Engels, S., Stenram, U., and Svanberg, S., Photody-namic therapy using intravenous delta-aminolaevulinic acid-induced protoporphyrin IX sensiti-sation in experimental hepatic tumours in rats, Br. J. Cancer, 74, 1526, 1996.

136. Mitchell, M.F.-L.G., Wright, T., Sarkar, A., Richards-Kortum, R., Hong, W.K., and Schottenfeld,D., Cervical human papillomavirus infection and intraepithelial neoplasia: a review, J. Natl. CancerInst. Monogr., 21, 17, 1996.

137. Mitchell, M.F., Hittelman, W.K., Lotan, R., Nishioka, K., Tortolero-Luna, G., Richards-Kortum,R., and Hong, W.K., Chemoprevention trials in the cervix: design, feasibility, and recruitment,J. Cell. Biochem., Suppl. 23, 104, 1995.

138. Ramanujam, N., Mitchell, M.F., Mahadevan, A., Warren, S., Thomsen, S., Silva, E., and Richards-Kortum, R., In vivo diagnosis of cervical intra epithelial neoplasia using 337-nm-excited laser-induced fluorescence, Proc. Natl. Acad. Sci. U.S.A., 91, 10193, 1994.

139. Ramanujam, N., Mitchell, M.F., Mahadevan, A., Thomsen, S., Silva, E., and Richards-Kortum, R.,Fluorescence spectroscopy: a diagnostic tool for cervical intra epithelial neoplasia (CIN), Gynecol.Oncol., 52, 31, 1994.

140. Barrett, N., Chronic peptic ulcer of the oesophagus, Br. J. Surg., 38, 175, 1950.141. Spechler, S.J. and Goyal, R.K., Barrett’s esophagus, New Engl. J. Med., 315, 362, 1987.142. Burbige, E.J. and Radigan, J.J., Characteristics of the columnar-lined (Barrett’s) esophagus,

Gastrointest. Endosc., 24, 133, 1979.143. Cameron, A.J., Sinmeister, A.R., and Ballard, D.J., Prevalence of columnar-lined (Barrett’s) esoph-

agus: comparison of population-based and autopsy findings, Gastroenterology, 99, 918, 1990.144. Spechler, S.J., Robbins, R.H., Rubins, H.B., Vincent, M.E., Heeren, T., Doos, W.G., Colton, T., and

Schimmel, E.M., Adenocarcinoma and Barrett’s esophagus, an overrated risk, Gastroenterology, 87,927, 1984.

145. Polopalle, S.C. and McCallum, R.W., Barrett’s esophagus: current assessment and future perspec-tives, Gastroenterol. Clin. North Am., 19, 733, 1990.

146. Vo-Dinh, T. and Mathur, P., Optical diagnostics and therapeutic technologies in pulmonary med-icine, in Interventional Bronchoscopy, Bollinger, D.T. and Mathur, P., Eds., Basel-Karger Publishers,Basel, 2000, p. 267.

147. Koenig, F., McGovern, F.J., Althausen, A.F., Deutsch, T.F., and Schomacker, K.T., Laser inducedautofluorescence diagnosis of bladder cancer, J. Urol., 156, 1597, 1996.

148. Anidjar, M., Ettori, D., Cussenot, O., Meria, P., Des Grandchamps, F., Cortesse, A., Teillac, P.,LeDuc, A., and Avrillier, S., Laser induced autofluorescence diagnosis of bladder tumors: depen-dence on the excitation wavelength, J. Urol., 156, 1590, 1996.

149. Schomacker, K.T., Frisoli, J.K., Compton, C.C., Flotte, T.J., Richter, J.M., Nishioka, N.S., andDeutsch, T.F., Ultraviolet laser-induced fluorescence of colonic tissue: basic biology and diagnosticpotential, Lasers Surg. Med., 12, 63, 1992.

150. Cothren, R.M., Sivak, M.V., VanDam, J., Petras, R.E., Fitzmaurice, M., Crawford, J.M., Wu, J.,Brennan, J.F., Rava, R.P., Manoharan, R., and Feld, M.S., Detection of dysplasia at colonoscopyusing laser-induced fluorescence: a blinded study, Gastrointest. Endosc., 44, 168, 1996.

©2003 CRC Press LLC

Page 50: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

151. Cothren, R.M., Richards-Kortum, R., Sivak, M.V., Fitzmaurice, M., Rava, R.P., Boyce, G.A., Dox-tader, M., Blackman, R., Ivanc, T.B., Hayes, G.B., Feld, M.S., and Petras, R.E., Gastrointestinaltissue diagnosis by laser-induced fluorescence spectroscopy at endoscopy, Gastrointest. Endosc., 36,105, 1990.

152. Lashner, B.A. and Brzezinski, A., Cancer mortality-rates in ulcerative-colitis surveillance programs,Gastroenterology, 106, 278, 1994.

153. Lashner, B.A., Provencher, K.S., Bozdech, J.M., and Brzezinski, A., Worsening risk for the devel-opment of dysplasia or cancer in patients with chronic ulcerative-colitis, Gastroenterology, 106,A718, 1994.

154. Jensen, P., Krogsgaard, M.R., Christiansen, J., Braendstrup, O., Johansen, A., and Olsen, J., Observervariability in the assessment of type and dysplasia of colorectal adenomas, analyzed using kappa-statistics, Dis. Colon Rectum, 38, 195, 1995.

155. Kolli, V.R., Shaha, A.R., Savage, H.E., Sacks, P.G., Casale, M.A., and Schantz, S.P., Native cellularfluorescence can identify changes in epithelial thickness in vivo in the upper aerodigestive tract,Am. J. Surg., 170, 495, 1995.

156. Kolli, V.R., Savage, H.E., Yao, T.J., and Schantz, S.P., Native cellular fluorescence of neoplastic upperaerodigestive mucosa, Arch. Otolaryngol.-Head Neck Surg., 121, 1287, 1995.

157. Dhingra, J.K., Perrault, D.F., McMillan, K., Rebeiz, E.E., Kabani, S., Manoharan, R., Itzkan, I., Feld,M.S., and Shapshay, S.M., Early diagnosis of upper aerodigestive tract cancer by autofluorescence,Arch. Otolaryngol.-Head Neck Surg., 122, 1181, 1996.

158. Fryen, A., Glanz, H., lohmann, W., Dreyer, T., and Bohle, R.M., Significance of autofluorescencefor the optical demarcation of field cancerisation in the upper aerodigestive tract, Acta Otolaryngol,(Stockholm), 117, 316, 1997.

159. Heintzelman, D.L., Utzinger, U., Fuchs, H., Zuluaga, A., Gossage, K., Gillenwater, A.M., Jacob, R.,Kemp, B., and Richards-Kortum, R.R., Optimal excitation wavelengths for in vivo detection of oralneoplasia using fluorescence spectroscopy, Photochem. Photobiol., 72, 103, 2000.

160. Zargi, M.S.L., Fajdiga, I., Bubnic, B., Lenarcic, J., and Oblak, P., Detection and localization of earlylaryngeal cancer with laser-induced fluorescence: preliminary report, Eur. Arch. Otorhinolaryngol.,Suppl. 1, S113, 1997.

161. Masters, B.R., So, P.T., and Gratton, E., Multiphoton excitation fluorescence microscopy andspectroscopy of in vivo human skin, Biophys. J., 72, 2405, 1997.

162. Leffell, D.J., Stetz, M.L., Milstone, L.M., and Deckelbaum, L.I., In vivo fluorescence of human skin.A potential marker of photoaging, Arch. Dermatol., 124, 1514, 1988.

163. Klinteberg, C., Enejder, A.M.K., Wang, I., Andersson, E., Svanberg, S., and Svanberg, K., Kineticfluorescence studies of 5-aminolaevulinic acid-induced protoporphyrin IX accumulation in basalcell carcinomas, J. Photochem. Photobiol., B 49, 120, 1999.

164. Chissov, V.I., Sokolov, V.V., Filonenko, E.V., Menenkov, V.D., Zharkova, N.N., Kozlov, D.N., Poli-vanov, I.N., Prokhorov, A.M., Pyhov, R.L., and Smirnov, V.V., Clinical fluorescent diagnosis oftumors using photosensitizer photogem., Khirurgiia (Moskva), 5, 37, 1995.

165. Heyerdahl, H., Wang, I., Liu, D.L., Berg, R., Andersson-Engels, S., Peng, Q., Moan, J., Svanberg,S., and Svanberg, K., Pharmacokinetic studies on 5-aminolevulinic acid-induced protoporphyrinIX accumulation in tumours and normal tissues, Cancer Lett., 112, 225, 1997.

166. Leunig, A., Rick, K., Stepp, H., Goetz, A., Baumgartner, R., and Feyh, J., Fluorescence photodetec-tion of neoplastic lesions in the oral cavity following topical application of 5-aminolevulinic acid,Laryngo-Rhino-Otologie, 75, 459, 1996.

167. von Holstein, C.S., Nilsson, A.M., Andersson-Engels, S., Willen, R., Walther, B., and Svanberg, K.,Detection of adenocarcinoma in Barrett’s oesophagus by means of laser induced fluorescence, Gut,39, 711, 1996.

©2003 CRC Press LLC

Page 51: Biomedical Diagnostics II: Optical Biopsy · 28.2.2.1 Molecular Electronic Energies In the study of luminescence, we are first concerned with determination of the electronic states

168. Baert, L., Berg, R., Vandamme, B., Dhallewin, M.A., Johansson, J., Svanberg, K., and Svanberg, S.,Clinical fluorescence diagnosis of human bladder-carcinoma following low-dose photofrin injec-tion, Urology, 41, 322, 1993.

169. Svanberg, K., Andersson, T., Killander, D., Wang, I., Stenram, U., Andersson-Engels, S., Berg, R.,Johansson, J., and Svanberg, S., Photodynamic therapy of nonmelanoma malignant tumors of theskin using topical delta-amino levulinic acid sensitization and laser irradiation, Br. J. Dermatol.,130, 743, 1994.

170. Sterenborg, H., Saarnak, A.E., Frank, R., and Motamedi, M., Evaluation of spectral correctiontechniques for fluorescence measurements on pigmented lesions in vivo, J. Photochem. Photobiol.,B-Biol., 35, 159, 1996.

171. Folli, S., Wagnières, G., Pelegrin, A., Calmes, J.M., Braichotte, D., Buchegger, F., Chalandon, Y.,Hardman, N., Heusser, C., Givel, J.C., Chapuis, G., Chatelain, A., Vandenbergh, H., and Mach, J.P.,Immunophotodiagnosis of colon carcinomas in patients injected with fluoresceinated chimericantibodies against carcinoembryonic antigen, Proc. Natl. Acad. Sci. U.S.A., 89, 7973, 1992.

©2003 CRC Press LLC