Open Access
12 January 2024 Photoacoustic imaging for cutaneous melanoma assessment: a comprehensive review
Author Affiliations +
Abstract

Significance

Cutaneous melanoma (CM) has a high morbidity and mortality rate, but it can be cured if the primary lesion is detected and treated at an early stage. Imaging techniques such as photoacoustic (PA) imaging (PAI) have been studied and implemented to aid in the detection and diagnosis of CM.

Aim

Provide an overview of different PAI systems and applications for the study of CM, including the determination of tumor depth/thickness, cancer-related angiogenesis, metastases to lymph nodes, circulating tumor cells (CTCs), virtual histology, and studies using exogenous contrast agents.

Approach

A systematic review and classification of different PAI configurations was conducted based on their specific applications for melanoma detection. This review encompasses animal and preclinical studies, offering insights into the future potential of PAI in melanoma diagnosis in the clinic.

Results

PAI holds great clinical potential as a noninvasive technique for melanoma detection and disease management. PA microscopy has predominantly been used to image and study angiogenesis surrounding tumors and provide information on tumor characteristics. Additionally, PA tomography, with its increased penetration depth, has demonstrated its ability to assess melanoma thickness. Both modalities have shown promise in detecting metastases to lymph nodes and CTCs, and an all-optical implementation has been developed to perform virtual histology analyses. Animal and human studies have successfully shown the capability of PAI to detect, visualize, classify, and stage CM.

Conclusions

PAI is a promising technique for assessing the status of the skin without a surgical procedure. The capability of the modality to image microvasculature, visualize tumor boundaries, detect metastases in lymph nodes, perform fast and label-free histology, and identify CTCs could aid in the early diagnosis and classification of CM, including determination of metastatic status. In addition, it could be useful for monitoring treatment efficacy noninvasively.

1.

Introduction

Over the last few decades, the global incidence of cutaneous melanoma (CM) has continued to rise.15 Melanoma is the fifth most common cancer in the United States, with high morbidity and mortality.6 While only 2% of skin cancers are diagnosed as melanoma, it accounts for 75% of skin cancer deaths annually.710 Melanoma tumor depth is an important prognostic factor, along with ulceration status.11 Early detection and diagnosis is critical.12 The 5-year survival rate is 99% for localized melanomas but decreases to 63% with regional metastases and 20% with distant metastases.8

Biopsies have long been considered the diagnostic standard for melanoma.13,14 Performing biopsies can result in pain, anxiety, scarring, and disfigurement for patients, as well as a considerable cost to the healthcare system. Given the high mortality associated with melanoma depth and metastasis, there is an urgent need for accurate, noninvasive methods to detect and monitor the disease. As a result, several noninvasive imaging techniques have been developed for skin imaging1517 including, among others, infrared imaging, hyperspectral/multispectral imaging, reflectance confocal microscopy (RCM), optical coherence tomography (OCT),1835 and photoacoustic (PA) imaging (PAI).3640 Many noninvasive imaging modalities have been used to study different aspects of CM, as comprehensively described in review articles.4143 Beyond their capabilities for an initial diagnosis, other opportunities for melanoma staging through imaging involve analysis of lymph nodes for metastases and detection of circulating (melanoma) tumor cells (CTCs).

PAI, also known as optoacoustic imaging, is an emerging noninvasive imaging modality4452 in which a nano-pulsed excitation light is absorbed by tissue chromophores (such as hemoglobin, lipids, bilirubin, and melanin) or exogenous contrast agents (such as organic dyes or nanoparticles) leading to a transient localized thermoelastic expansion.51,5358 This induces the generation of PA signals, which are then detected by an ultrasound (US) transducer and reconstructed into a two-dimensional (2D) or three-dimensional (3D) image. The strength of the PA signals correlates with tissue chromophore absorption properties, which depend on the wavelength and the chromophore’s absorption spectrum. The combination of using light illumination and US detection provides PAI with advantages (e.g., high sensitivity and specificity at a greater imaging depth) over other modalities that utilize only light or US for signal generation and detection.39,59 Optical imaging modalities, such as RCM and OCT, have shown the ability to differentiate tissue microstructures; however, they have limited penetration depth due to the scattering of light in tissue,6062 even with performing enhancement postprocessing algorithms.18,20,6379 US imaging provides improved penetration depth80 but has limited ability to differentiate between skin conditions.81

PAI can be classified by axial resolution of generated images into PA macroscopy, mesoscopy, and microscopy. Macroscopy utilizes lower US frequencies, which allows for greater penetration depths, in the range of a few centimeters.8284 This has enabled noninvasive imaging of breast cancer,85 Crohn’s disease activity,86 brown fat metabolism,87 and various blood vessels located in the neck (carotid),88 hands,89 and feet,90 as well as whole-body imaging in animals.91,92 Microscopy utilizes higher US frequencies and allows for imaging with resolution capable of imaging individual cells9396 but with much lower penetration depth. Mesoscopy is a bridge between macroscopy and microscopy, utilizing US frequencies that image at penetration depths of several millimeters,82 making it optimal for dermatological investigations.97 Mesoscopy has been used to visualize: skin morphology generally,87,98,99 vascular patterns, morphology in psoriasis and atopic dermatitis,100 nailbed microvasculature,101 and vasodilation induced by hyperthermia.102

Another way to classify PAI systems is based on how the image is reconstructed (Fig. 1): in PA tomography (PAT), also known as PA computed tomography (PACT), a reconstruction algorithm is used to convert the PA signals collected at different locations/angles around the object into an image,47,48,103,104 whereas in raster scan PAI, the pixels/voxels of the image are generated as the sample is raster scanned. Based on the arrangement of transducers, PAT can further be categorized into single element PAT, linear array PAT (LA-PAT), ring array PAT, and hemispherical PAT,47,48,103,105,106 but LA-PAT is more widely implemented than other forms. Raster scan PAI has also been implemented in a variety of configurations,47,107109 which can be divided into optical resolution PA microscopy (OR-PAM) and acoustic resolution PA microscopy (AR-PAM). In OR-PAM, the optical beam is focused to a tight spot, smaller than the acoustic detection sensitivity area and raster scanned point by point using an optical scanner.110112 With OR-PAM, reaching to the cellular level resolution is achievable; however, the imaging depth is limited to a few millimeters.108 In AR-PAM, a spherically focused transducer scans the sample.110,113,114 This yields a greater penetration depth than OR-PAM but with a coarser resolution. Raster scanning optoacoustic mesoscopy (RSOM) is an implementation of AR-PAM that has also been used for skin imaging. Because raster scanning images samples point by point, image acquisition can be slow. Configurations for different PA systems (PACT, RSOM, OR-PAM, and AR-PAM) are shown in Fig. 1.

Fig. 1

Different PAI configurations used for melanoma analysis: (a) PACT, (b) AR-PAM, and (c) OR-PAM (inset: RSOM). PACT, photoacoustic computed tomography; AR-PAM, acoustic-resolution photoacoustic microscopy; OR-PAM, optical-resolution photoacoustic microscopy; RSOM, raster scanning optoacoustic mesoscopy.

JBO_29_S1_S11518_f001.png

A third way to categorize PAI systems is whether they use a single wavelength laser to collect images or multiple wavelengths. In general, a single wavelength cannot distinguish between different chromophores.92 In multispectral PAI (MPAI), two or more wavelengths92,115119 are used to differentiate between chromophores. For instance, melanin and hemoglobin can be differentiated one from another using 584 and 764 nm.115,116,120 MPAI has been used to study numerous diseases, including prostate cancer,121 glioblastoma,122,123 ovarian cancer,124 thyroid disease,125,126 Crohn’s disease,86 liver disease,127 cardiac injury,128 systemic sclerosis,129 and cutaneous tumors.130,131

In terms of applications, PAI can also be categorized with regards to the type of questions that the imaging modality can be used to answer, namely, fitness of each PAI system to determine: (a) melanoma detection and depth measurement, (b) tumor angiogenesis, (c) lymph node metastases, (d) CTCs, and (e) virtual histology (see Fig. 2). Exogenous contrast agents have also been applied for some of these applications. Hardware designs of selected PAI systems that have been used in some of these applications are shown in Fig. 3. This categorization is consistent with the organization of the paper.

Fig. 2

PAI applications for characterizing melanoma, discussed in this review. CTC, circulating tumor cells; PAI, photoacoustic imaging.

JBO_29_S1_S11518_f002.png

Fig. 3

Hardware designs of selected PAI systems. (a) For melanoma detection and depth measurement: (i) all optical OR-PAM combined with OCT system. SLD, super luminescent diode; PD, photodetector; FC, fiber coupler; G, grating; GM, scanning galvanometer; C1, C2, and C3, collimators; L1 and L2: lenses; M, mirror; DM, dichroic mirror; FPGA, field programmable gate array; ADC, analog-to-digital converter; OL, objective lens; S, sample; OCT, optical coherence tomography; AOPA, all-optical integrated photoacoustic. Reproduced from Ref. 132. (ii) Multiwavelength AR-PAM system. Reproduced from Ref. 113. (iii) 3D wide-field multispectral photoacoustic imaging of human melanomas, a dual-modal photoacoustic and US imaging system. Reproduced from Ref. 133. (iv) Multiscale PAM. NDF, neutral density filter; DM, dichroic mirror; M1 and M2, mirrors; AD, achromatic doublet; P, prism; AL, acoustic lens; UT, US transducer; PS, polygon-scanning mirror, DAQ, data acquisition. Reproduced from Ref. 134. (v) Handheld photoacoustic system for melanoma imaging. Reproduced from Ref. 135. (b) Melanoma detection and angiogenesis: (i) multiscale PAM. NDF, neutral density filter; DM, dichroic mirror; M1 and M2, mirrors; AD, achromatic doublet; P, prism; AL, acoustic lens; UT, US transducer; PS, polygon-scanning mirror, DAQ, data acquisition. Reproduced from Ref. 134. (ii) Schematic of faster raster scanning optoacoustic mesoscopy (RSOM). Reproduced from Ref. 109. (iii) Multiwavelength OR-PAM system. OL, objective lens; NDF, neutral dispersion fiber; MLPF, motorized long-pass filter; L1 and L2, achromatic lenses; CL, corrective lens; FT, focused transducer; WD, water dish; DAQ, data acquisition; OAC, optical/acoustic combiner. Reproduced from Ref. 136. (iv) Multispectral LA-PAT system. OPO, optical parameter oscillator. Reproduced from Ref. 115. (c) Flow cytometry: (i) single wavelength photoacoustic flow cytometry. Reproduced from Ref. 137. (ii) Multispectral photoacoustic flow cytometry. ADC, analog-to-digital converter. Reproduced from Ref. 138. (iii) Single-impulse panoramic photoacoustic computed tomography. Reproduced from Ref. 139. (iv) Optical resolution photoacoustic flow cytometry system. PD, photodiode. Reproduced from Ref. 140. (d) Metastasis to lymph nodes: (i) photograph and schematic of dual-modal photoacoustic and ultrasound imaging system with a solid-state dye laser. US, ultrasound; Tx, transmit; Rx, receive; TR, transducer; WT, water tank; ED, engineered diffuser; SD, solid-state dye. Reproduced from Ref. 141. (c) Multispectral optoacoustic tomography system with three different transducers used in the study, inVision 128, 2D handheld and 3D handheld. Reproduced from Ref. 117. (e) Virtual histology: system diagram of combined UV-PARS and UV scattering microscopy system. M, mirror; L, lens; BS, beamsplitter; HBS, harmonic beamsplitter; RO, reflective objective; PD, photodiode. Reproduced from Ref. 142. (f) Exogenous contrast agents: types of contrast agents utilized, implemented in animal studies. Reproduced from Refs. 143 and 144.

JBO_29_S1_S11518_f003.png

2.

Methods

Several reviews have been written on skin imaging applications of PAI,45,46,91,145164 but only one, that we are aware of, on melanoma imaging applications of PAI, and it is restricted to PAT applications and was published several years ago.158 We therefore conducted a literature review of primary research describing PAM and PAT to investigate CM that were published before December 2023. The literature search was conducted using the terms “PA” and “optoacoustic” with “melanoma.” Studies were excluded if they (1) did not utilize PA or optoacoustic methods, (2) were on cancers other than CM, (3) were not in English, (4) were not primary research papers, or (5) did not describe the parameters of the PA system utilized in the study. Our primary source was Google Scholar, which yielded 12,300 total results. Of the 12,300 results, 102 studies103,109,113,115117,132140,165245 met our criteria and are reviewed here.

3.

Investigations of CM Using PAI

3.1.

Melanoma Detection and Depth Measurement

Imaging techniques able to detect melanoma with high sensitivity and specificity could greatly reduce the number of biopsies that are currently performed. Once a melanoma is confirmed through detection, the next step is melanoma staging. Staging melanoma is critical since it determines prognosis and treatment options.12 Melanoma depth (Breslow depth) is one of the two variables used to stage localized melanoma;12 the other one is ulceration status. The tumor (T) category of the tumor-node-metastasis staging system depends on tumor depth and is classified as T1 (1.00  mm), T2 (1.01 to 2.00 mm), T3 (2.01 to 4.00 mm), or T4 (>4.00  mm).12 For T1 melanomas, the 10-year survival rate is 92%, but drops to 50% for T4 melanomas.12 Excisional biopsy is required for staging lesions to accurately determine the Breslow depth.11,246250 Imaging modalities could be used to stage melanoma, guide surgical planning, and prevent incomplete excisions and subsequent additional surgeries.

3.1.1.

Animal studies

Oh et al.116 used dual-wavelength AR-PAM to detect melanoma in mice inoculated with highly invasive B16 skin melanoma cells. Due to the difference in peak optical absorption between melanin and hemoglobin, a near-infrared (NIR) light (764 nm) and a visible light (584 nm) were used to visualize the melanoma [see Fig. 4(a)(i)] and surrounding vascular structures [see Fig. 4(a)(ii)]. Figures 4(a)(iii) and 4(a)(iv) illustrate the B-scan images across the red dotted line in Figs. 4(a)(i) and 4(a)(ii). The maximum thickness of the melanoma was found to be 0.5 mm. Zhou et al.187,188 conducted several experiments to study PAI in nude mice using a similar melanoma model. They used a handheld AR-PAM to measure the depth of melanoma.188 Their PAM utilized an “annular-shaped” light illumination method, in which light (with 8 mm inner diameter and 20 mm outer diameter) bypasses the center of the tumor and instead is delivered in the direction normal to the surface.188 The melanoma depth (3.66 mm) as measured by their system [see Fig. 4(b)] corresponded well to the actual, postexcisional thickness (3.75 mm).188 In another experiment utilizing a LA-PAT system, tumor depth and volume were measured and revealed an increase in tumor depth and volume from day 3 [Fig. 4(c)(i)] to day 6 [Fig. 4(c)(ii)] after injection with the B16 cells.187 The depth increased from 1.32 to 2.77 mm and volume increased from 22.365 to 71.931  mm3. Moreover, the system was able to measure the rate of growth for both depth and volume of tumor. Recently, Wang et al.184 built a hybrid PA/US system with a sound-light coaxial/confocal design by punching a 2 mm diameter hole in the center of the transducer to deliver the laser light. Melanomas in mice were imaged in vivo at day 7 [see Fig. 4(d)Ii)] and day 30 [see Fig. 4(d)(ii)] after B16 cell injection. A clear growth in tumor size and depth is observed as shown in B-scan images of melanoma at day 7 [see Fig. 4(d)(iii)] and day 30 [see Fig. 4(d)(iv)]. Moreover, PAI visualized microvasculature around the tumor. Another experiment with mice was conducted by Wang et al.251 using a dual-wavelength AR-PAM with visible and NIR light combined with US to image sub-CM. The fused images from the two wavelengths enrich the imaging information and allow more accurate detection of the melanoma, differentiating it from normal tissue. With the 3D distribution the boundary detection of the melanoma is easier and accurate and could be further enhanced by the US structural information helping the identification of the tissue boundaries and precisely locating the sub-CM [see Figs. 4(e)(i)4(e)(iii)].

Fig. 4

Melanoma detection and depth measurement in live animal models using different PAI configurations. (a) PA images of melanoma and vascular distribution in nude mouse skin. (i), (ii) En face PA images for the NIR light source (λ=764  nm) and visible light source (λ=584  nm) respectively: 1, melanoma; 2, vessels perpendicular to imaging plane; 3, vessels horizontal to imaging plane; 4, skin. (iii), (iv) B-Scan PA images along the red line in panels (i) and (ii). Reproduced from Ref. 116. (b) PA image of the melanoma showing both the top and bottom boundaries in nude mice in vivo. The red dots outline the melanoma. Reproduced from Ref. 188. (c) LA-PAT images of melanoma acquired in nude mice on day 3 (i) and day 6 (ii) after tumor implantation. Reproduced from Ref. 187. (d) PA maximum amplitude projection (MAP) images of melanoma in mice at (i) day 7, (ii) day 30 and PA B-scan images on (iii) day 7 and (iv) day 30. Reproduced from Ref. 184. (e) Fused 3D visible light and NIR PA MAP images in mice with melanoma. (i) visible light and US, (ii) NIR light and US, and (iii) visible and NIR light. Reproduced from Ref. 251.

JBO_29_S1_S11518_f004.png

3.1.2.

Human studies

In a pilot study, Zhou et al.135 analyzed 10 melanomas in 7 patients using a LA-PAT system [see Fig. 5(a)] immediately preceding excisional biopsy. Melanomas at depths from 0.2 to 6 mm were visualized, but deeper tumors were beyond the detection limit of the LA-PAT system. Figure 5(a) shows the resultant PA images [see Figs. 5(a)(i) and 5(a)(iv)], photographs [see Fig. 5(a)(ii)5(a)(v)], and histological images [see Figs. 5(a)(iii) and 5(a)(vi)] of two patients, one with CM metastases on the left lower extremity and the other with primary acral lentiginous melanoma on the right foot. Histological images acquired from both patients showed that the detected melanoma depth was consistent with actual Breslow depth, showing the promising capability of the system for detection. Kim et al.177 developed an integrated PA and US imaging (PAUSI) system by combining a clinical US machine and a multispectral portable laser. The imaging system was utilized to image a patient’s melanoma ex vivo after excision [see Fig. 5(b)(i)]. The amplitude of the PA signal corresponds to the amount of melanocytes in the local area. The PA signals in melanoma, indicated by the white triangles [see Fig. 5(b)(ii)], were predominantly generated by optical wavelengths of 800 and 1064 nm. In contrast, the PA signals from the marking pen regions indicated by the white arrows were predominantly generated from the 680-nm laser. Moreover, PA imaging indicated areas of melanoma that are not visible to the surgeon [yellow triangles in Fig. 5(b)(ii)]. In addition, the measured thickness of the melanoma region (420±320  μm) matched well with the histopathological results. A larger study on PAI was conducted by Breathnach et al.115 using LA-PAT and spectral unmixing, to pre-operatively image 32 pigmented lesions suspicious for melanoma in 27 patients. With spectral unmixing, they separated the absorption signature of melanin-containing cells and mapped the spatial distribution of it based on this signature, because the absorption spectrum of melanin varies within the NIR region. The lesion depths measured by PA were highly correlated with histopathologic measurements, with a correlation coefficient of 0.98 for benign lesions and 0.99 for melanomas. Using the same PA probe, the lesion architecture, adnexal depth (depth of lesion extension into the skin appendages), and various skin layers were also imaged, allowing for differentiation of superficial from invasive lesions based on their dermal-epidermal junction penetration, i.e., lesion penetrated through the dermal-epidermal junction [see Fig. 5(c)]. According to the authors, due to the tissue sample dehydration and loss of skin tension in vivo, PAI overestimated lesion depth as compared with histopathology. Recently, Park et al.133 utilized a 3D multispectral PAT system to noninvasively measure depth and outline the boundary of melanomas for optimal surgical margin selection. Six melanoma patients were examined. They imaged melanoma of various forms, sizes (1.3 to 30 mm for lateral diameter and 0.6 to 9.1 mm for depth), and locations (sole, chest, thigh, heel, and palm) using their multispectral PA/US system [see Fig. 5(d)]. For five of the six case studies, melanoma depth was measured using multispectral analysis and confirmed a high correlation against histopathologic results with a mean absolute error of 0.36 mm. In a signal-based study, Swearingen et al.252 investigated if label-free MPAI could distinguish vascular from pigmented (melanotic) lesions in 15 human patients. Excitation lights at 422 and 530 nm were used. At 422 nm, melanotic lesion showed a lower PA signal [see Fig. 5(e)(i)] compared with vascular lesion [see Fig. 5(e)(ii)]. Similarly, at 532 nm, melanotic lesion showed higher PA signal [see Fig. 5(e)(iii)] compared with vascular lesion [see Fig. 4(e)(iv)]. The experiment proved the ability of MPAI to distinguish between vascular and pigmented lesions. About 15 lesions were biopsied after imaging, revealing 8 vascular and 7 pigmented lesions. Data analysis was carried out via two statistical methods, the classical method (standard multivariate analysis classification techniques) and a Bayesian-model-based approach. The classical method attained a perfect lesion diagnosis rate, whereas the Bayesian approach had a 20% error rate.

Fig. 5

Melanoma detection and depth measurement in human studies using different PAI configurations. (a) PAT of melanoma of two patients: (i) melanoma image acquired with a PA depth of 1.9 mm (cPAdepth=1.67  mm); (ii) cutaneous melanoma metastasis in a patient lower leg; (iii) histology of the excised melanoma, showing actual Breslow depth of 1.67 mm; (iv) PAT melanoma image of the acral lentiginous melanoma with PA depth of 0.70 mm (cPAdepth=0.62  mm); (v) acral lentiginous melanoma (data not shown: pBD=0.48  mm); and (vi) histology after complete excision, with actual BD of 0.78 mm. BD, Breslow depth; cPA, corrected photoacoustic depth; PAT, photoacoustic tomography; pBD, provisional Breslow depth. Reproduced from Ref. 135. (b) (i) Photograph and (ii) ex vivo PA image of excised melanoma tissue from a male patient. The melanoma regions are represented by dark red to bright-yellow color (white triangles), and the marking-pen regions are represented by dark green to bright-green color (white arrows). Yellow arrows indicate possible melanomas not found by histology. Reproduced with permission from Ref. 177. (c) PA image of in situ melanoma on upper left extremity on a patient using LA-PAT. Reproduced with permission from Ref. 115. (d) Measurement of PA depth of a nodular type of melanoma. (i) Photoacoustic MAP; (ii) photoacoustic MAP, overlaid with US image; and (iii)–(iv) photoacoustic unmixed and photoacoustic unmixed overlaid with US images, respectively. Blue arrows indicate the melanoma invasion and yellow arrows are the bottom boundary of the melanoma. MAP, maximum amplitude projection. Reproduced from Ref. 133. (e) Representative PA signals from human subjects with either (i), (iii) pigmented and (ii), (iv) vascular lesions at 422 and 530 nm, respectively. Reproduced with permission from Ref. 252.

JBO_29_S1_S11518_f005.png

Figures 4 and 5 and the related text describe results from a selection of PAI studies of melanoma detection and depth measurement. Table 1 includes specific characteristics of the PAI systems used in published studies on melanoma detection/depth measurement for both animal and human studies.

Table 1

Specific characteristics of PAI systems used in published studies on melanoma detection/depth measurement.

PAI modalityWavelengthsUS transducerResolutionImaging modelStudy result
AR-PAMλ1: 764 nmCF: 50 MHzLR: 45  μmMice (B16 subcutaneous injection)116Dual wavelength melanoma imaging and depth calculation
λ2: 584 nmBW: 70%AR: 15  μm
Melanoma depth:
NE: 1λ1: 2D: 0.3 mm and 3D: 0.5 mm
λ2: 2D: 0.45 mm and 3D: 0.15 mm
AR-PAMλ: 650 nmCF: 25 MHzLR: 230  μmMice (B16 subcutaneous injection)188Annular-shaped light illumination method for melanoma detection
BW: 100%AR: 59  μm
NE: 1
Melanoma depth: 3.66 mm
AR-PAMλ1: 422 nmCF: 25 MHzNLHuman subjects with melanoma252Study and classification of 15 human subjects with eight vascular and seven pigmented lesions
λ2: 530 nmBW: 100%
NE: 1
AR-PAMλ1: 532 nmCF: 25 MHzLR: 0.17 mmMice (B16 subcutaneous injection)251Detection of melanoma with a combination of multi-wavelength PA images and US
λ2: 1064 nmBW: 80%AR: 0.12 mm
NE: 1
OR-PAMλ: 532 nmCF: 29 MHzLR: 3.5  μmMice (B16 subcutaneous injection)184Characterization of tumor vasculature and depth
BW: 80%AR: 60  μm
NE: 1Melanoma depth: 2.58 mm
OR-PAMλ: 584 nmCF: 100 MHzLR: 5  μmMice (B16 implanted in ear)111Differentiation of blood vessels and melanoma without contrast agent after 3 and 7 days of implantation
AR: 15  μm
BW: NL
NE: 1
OR-PAMλ: 532 nmCF: 125 MHzLR: 0.8  μmMelanoma cells fixed in formalin210Detection and imaging of melanoma cells with high resolution ex vivo
BW: 80%AR: 7.6  μm
NE: 1
OR-PAMλ: 1064 nmCF: 41 MHzLR: 36  μmMice (B16 subcutaneous injection)220Detection of melanoma without exogenous contrast agent
BW=NLAR: 61  μm
OR-PAMλ: 532 nmCF: 13 MHzLR: 8.5  μmMice (B16 implanted in ear)231Detection of the boundaries of melanoma and depth profiling
BW: 60%AR: 150  μm
NE: 1
OR-PAMλ: 600 to 1000 nmCF: 25 MHzNLMice (B16 subcutaneous injection to torso and ear)136Differentiating melanomas from blood vessels and monitoring melanoma growth
BW: NL
NE: 1
All optical OR-PAMλ: 532 nmMichelson detectorLR: 13  μmMice (B16 implanted in ear)132,232Detection and monitoring of melanoma and angiogenesis with dual modality (PAM/OCT)
AR: 20  μm
SW-PAMλ: 532 nmCF: 40 MHzLR: 400 nmMice (B16 implanted in ear)205Detection of melanoma and visualization of nearby vasculature. Monitoring melanoma growth over 4 days
NA: 0.5
NE: 1
LA-PATλ: 680 nmCF: 21 MHzLR: 119  μmMice (B16 subcutaneous injection)187Detection of melanoma depth and volume
BW: 55%AR: 86  μm
NE: 256
LA-PATλ: 700 nmCF: 45 MHzLR: 8  μmMice (B16 subcutaneous injection)237Monitoring melanoma volume after photodynamic treatment
BW: 55%AR: 15  μm
NE: 256Melanoma depth: 4 mm
LA-PATλ: 680 nmCF: 21 MHzLR: 119  μmHuman subjects with melanoma135Detection of melanomas with depths of 0.2 to 6 mm
F: 10  mJ/cm2BW: 70%AR: 86  μm
NE: 256The detection limit was 10 mm
LA-PATλ1: 800 nmCF: NLLR: 1200  μmHuman subjects with melanoma177Detection of melanoma and measurement of thickness of the melanoma region
λ2: 1064 nmBW: NLAR: 205  μm
λ3: 680 nmNE: 128
Melanoma depth: 420±320  μm
LA-PATλ1: 680 nmCF: 40 MHzNLHuman subjects with melanoma115Testing feasibility of LA-PAT probe and measuring tumor depth. High correlation with histopathology (0.99 for melanomas, 0.98 for benign lesions)
λ2: 700 nmBW: 55%
λ3: 750 nmNE: 256
λ4: 850 nm
λ5: 900 nm
LA-PATλ: 700 nmCF: 40 MHzLR: 140  μmHuman subjects with melanoma218Detection of melanoma and identification and measurement of lesion boundaries
λ: 850 nmNE: 256
LA-PATλ: 680 to 970 nmCF1: 20 MHzLR1: 110  μmHuman subjects with melanoma (ex vivo samples)103Feasibility of PAI for noninvasive delineation of the borders of melanoma
BW1: 55%AR1: 50  μm
CF2: 30 MHzLR2: 165  μm
BW2: 80%
AR2: 75  μm
PATλ1=700  nmCF: 7.5 MHzLR: 1.0 mmHuman subjects with melanoma133Measurement of melanoma depth and confirmation of metastastatic melanoma
λ2=756  nmBW: 120%AR: 0.2 mm
λ3=796  nm
λ4=866  nmNE: 128Melanoma depth: 9.1 mm
λ5=900  nm
Note: λ, wavelength; F, fluence; CF, central frequency; BW, bandwidth; NE, number of elements; NL, not listed; IA, incident angle; LR, lateral resolution; AR, axial resolution; LA-PAT, linear array photoacoustic tomography; AR-PAM, acoustic resolution photoacoustic microscopy; OR-PAM, optical resolution photoacoustic microscopy; SW-PAM, subwavelength resolution photoacoustic microscopy.

3.2.

Measurement of Tumor Angiogenesis in Melanoma

Tumor angiogenesis refers to the formation of new blood vessels within a tumor, or the growth of blood vessels between a tumor and its surrounding tissues. Tumor-associated vasculature not only regulates the supply of nutrients and oxygen to the tumor but also expedites tumor invasion and metastasis. Therefore, as an essential indicator of disease progression, vascularization within a tumor can be used to assess the potential for metastasis.184,253 PAI has shown great promise in monitoring the progression of tumor angiogenesis143,254 using OR-PAM and also RSOM in mice and humans (Table 2 and Fig. 6).

Table 2

Summary of PAI studies on melanoma tumor angiogenesis.

PAI modalityLight sourceUS transducerResolutionImaging modelStudy result
RSOMλ: 532 nmCF1: 50 MHzLR: 18  μmMice (B16 subcutaneous injection to mammary pad)182Studying changes in the vascular network caused by melanoma
BW1: 160%AR: 4  μm
CF2: 100 MHz
BW2: 160%50 MHz: superior imaging of larger structures
100 MHz: better visualization of tumor microvasculature
All optical PAM/RCMλ: 532 nmLow-coherence interferometer:LR: 4.5  μmMice (B16 implanted in ear)186Dual modality system
PAM: visualized vascularity and pigmentation
CW: 1310 nm
SB: 45 nmRCM: illustrated cytological features
OR-PAMλ1: 570 nmCF: 75 MHzNLMice (B16 implanted in ear)185Detection of changes in tumor vascularity from 9 to 15 days after tumor inoculation
λ2: 1064 nmBW: NL
OR-PAMλ: 610 nmPVDFLR: 6  μmMice (B16 implanted in ear)234Detection of neovasculature surrounding the tumor 9 days after melanoma inoculation
CF: 25 MHzAR: 53  μm
OR-PAMλ1: 730 nmCF1: 7.5 MHzLR: 102  μmMice (B16 subcutaneous injection)241Quantification of the angiogenesis and melanoma thickness
λ2: 756 nmCF2: 31.5 MHzAR: 81  μm
λ3: 778 nm
λ4: 796 nm
λ5: 818 nm
OR-PAMλ1: 528 nmCF: 50 MHzLR: 7.8  μmMice (B16 implanted in ear)233Identification of the vasculature around the tumor with high resolution
λ2: 558 nmBW: 76%AR: 41  μm
NE: 1
OR-PAMλ1: 532 nmCF: 50 MHzLR OR: 7.1  μmMice (B16 subcutaneous injection to thigh)134Rapid visualization of melanoma boundaries. Imaging of the blood vasculature around the melanoma
λ2: 1064 nmBW: 70%LR AR: 112  μm
NE: 1AR: 10  μm
AR-PAMλ1: 584 nmCF: 50 MHzLR: 45  μmMice (B16 subcutaneous injection)113Identification of melanoma and angiogenesis surrounding the tumor
λ2: 764 nmBW: 70%AR: 15  μm
RSOMλ: 532 nmCF: 25 MHzNLHumans with melanoma109Quantifiable biomarkers were extracted from the vascular images by comparing benign nevi and melanomas
BW: 120%
CF: 80 MHz
BW: 100%
PARSλ: 532 nmLow coherence interrogation laserLR: 2.7  μmChicken eggs with melanoma227Study of superficial microvasculature with high lateral resolution
CW: 1310 nm
CL: 40  μm
Note: λ, wavelength; F, fluence; CF, central frequency; CW, central wavelength; SB, spectral bandwidth; CL, coherence length; NL, not listed; RSOM, raster scanning optoacoustic mesoscopy; OR-PAM, optical resolution photoacoustic microscopy; RCM, reflectance confocal microscopy; PARS, photoacoustic remote sensing microscopy.

Fig. 6

Angiogenesis studies in melanoma identification. (a) PA imaging of melanoma tumor vasculature in mice using (i) 50 MHz and (ii) 100 MHz detector. Reproduced with permission from Ref. 182. (b) PA images showing tumor growth over time. PA images of the tumor area acquired at (i) day 2 (D02), (ii) day 4 (D04), (iii) day 7 (D07), and (iv) day 9 (D09) after injection of melanoma cells, the thick arrow points to the tumor. The inset in every image shows the same region, from an area very close to the tumor, and shows two large vessels (the inset arrow points to that region), between which small vessels grow over time. Reproduced with permission from Ref. 182. (c) PA vascular imaging of melanoma in mouse at (i) day 9, (ii) day 13, (iii) day 14, and (iv) day 15. Reproduced with permission from Ref. 185. (d) PA images of human melanoma in vivo for three scanned regions: scan 1: inside the lesion, scan 2: boundary of the lesion, and scan 3: outside the lesion. (i)–(iii) Maximum amplitude projection (MAP) cross-sectional faster raster-scan optoacoustic microscopic (FRSOM) images. (iv)–(vi) MAP images in the coronal direction corresponding to the epidermal layer in panels (i)–(iii). (vii)–(ix) Coronal images corresponding to the dermal layer. EP, epidermal; DR, dermal. Reproduced with permission from Ref. 109. (e) In vivo OR and AR images of melanoma in mouse using multiscale PAM. (i) Top and side view MAP images obtained by OR-PAM at 532 nm. (ii) Top and side view MAP images obtained by AR-PAM at 1064 nm. (iii) Merged images of top and side view from OR and AR-PAM. Reproduced with permission from Ref. 134. OR, optical resolution; AR, acoustic resolution.

JBO_29_S1_S11518_f006.png

3.2.1.

Animal studies

In one study, Omar et al.182 utilized RSOM to visualize angiogenesis and tumor growth in melanomas of mice in vivo over several days. B16F10 melanoma cells were injected subcutaneously into the mammary fat pad of Hsd: Athymic Nude-Foxn1nu mice, and tumor growth and angiogenesis were monitored using spherically focused US detectors with central frequencies of 50 and 100 MHz. The 50 MHz detector was superior in imaging larger structures such as larger, oblique blood vessels [see Fig. 6(a)(i)], whereas the 100 MHz detector provided better visualization of tumor [appearing as a black hole in Fig. 6(a)(ii)] and newly sprouting vessels [see Fig. 6(a)(ii)]. Using the 50 MHz detector, tumor growth was recorded at day 2 [see Fig. 6(b)(i)], day 4 [see Fig. 6(b)(ii)], day 7 [see Fig. 6(b)(iii)], and day 9 [see Fig. 6(b)(iv)]. Each subfigure in Fig. 6(b) includes an inset taken from the proximity of the tumor. The growth of the tumor from day 2 to day 9 was illustrated by the growth of the black nonvascularized spot, indicated by a thick arrow in all the subfigures in Fig. 6(b). Moreover, in each inset, it is observed that upon interaction with the tumor, the two big vessels (denoted by a thinner arrow) start re-arranging; at the same time, smaller vessels start growing in that region, clearly representing tumor angiogenesis at a microvasculature level. Zhao et al.185 utilized OR-PAM at two wavelengths (570 and 1064 nm) to study melanoma tumor angiogenesis. B16 melanoma cells were injected subcutaneously into the mouse ear and tumors were imaged on day 9 [see Fig. 6(c)(i)], day 13 [see Fig. 6(c)(ii)], day 14 [see Fig. 6(c)(iii)], and day 15 [see Fig. 6(c)(iv)] after tumor inoculation in two mice. In the earlier day postinoculation, small diameter (<25 to 30  μm) vessels were most prevalent. On the following 2 days, the number of large diameter vessels (50 to 95  μm) increased while the proportion of small diameter vessels decreased. On day 15, vessels with diameter >100  μm were visualized. Moreover, the vessel density, vessel tortuosity, and fractional dimension (quantitative parameters they used to assess tumor growth) also showed an overall upward trend from day 9 to day 15. Thus, the authors concluded that with the growth of melanoma, the vascular networks become stronger and complex, which was consistent with visual results in Fig. 6(c). Another study by Zhou et al.186 used a combined all optical PA microscopy system and RCM system to study tumor growth and angiogenesis in mice who received subcutaneous injections of B16 melanoma cells in the ear. Their PAM showed irregular and linear vascular patterns, likely representing neovascularization of the dermis. The RCM enface image illustrated widespread pagetoid cells with cytologic atypia and nucleated cells within the dermal papilla. The PAM provided significant contrast and penetration depth, based on optical absorption properties, and visualized vascularity and pigmentation, whereas RCM illustrated cytological features. On the other hand, Xu et al.134 developed an integrated OR/AR-PAM system for multiscale imaging capability with high-speed wide-field imaging based on a polygon scanner. The polygon has six aluminum-coated surfaces that reflect the light and the acoustic beams: in this way six repeated cross-sectional scans can be obtained in one rotation of the motor, increasing image acquisition speed. With the two modalities and using two different wavelengths (532 and 1064 nm) a sub-CM is detected and separated from the surrounding microvasculature [see Fig. 6(e)].

3.2.2.

Human studies

Recently, angiogenesis of human melanomas has been imaged by He et al.109 using a single-breath-hold faster RSOM system to visualize the microvasculature of pigmented melanocytic lesion [see Fig. 6(d)]. The images from three different regions: scan 1 inside the lesion [see Figs. 6(d)(i), 6(d)(iv), and 6(d)(vii)], scan 2 in lesion boundary [see Figs. 6(d)(ii), 6(d)(v), and 6(d)(viii)], and scan 3 outside lesion [see Figs. 6d(iii), 6(d)(vi), and 6(d)(ix)], showed a difference on the epidermis and dermis vasculature patterns inside and outside the lesion. From the scans of 10 dysplastic nevi and 10 melanomas, a quantification of the vasculature features was performed to identify biomarkers for melanoma detection. A total of six biomarkers were calculated: the total blood volume, vessel density, average vessel length, tortuosity, fractal number, and lacunarity. It was found that those markers showed differences between malignant and benign lesions, supporting the possibility to use this system to improve melanoma diagnosis.

3.3.

Lymph Node Metastases

Melanoma survival rates are high when the disease is caught early, but decrease significantly after nodal or distant metastasis. The sentinel lymph node (SLN) is the hypothetical first lymph node draining the cancer; SLN metastasis has been shown to predict the pathologic state of the nodal basin.11 An SLN biopsy is typically performed during wide local excision if the Breslow depth is >1  mm; moreover, SLN biopsies are considered in ulcerated tumors of any depth.11,255 The SLN for a specific melanoma is identified through pre-operative lymphoscintigraphy, or intra-operatively using either a blue dye injection near the primary tumor site or a gamma probe with technetium-99 sulfur colloid.11 After excision, SLNs are examined via histopathology and immunohistochemistry for metastasis. However, these examinations can miss the presence of metastases due to false negative rates varying from 5% to 21%.256 Once a lymph node is removed, typically 6 to 10 sections of 6  μm thickness are taken and examined for metastasis. Thus, in a typical node with 1 cm length, there are hundreds of possible sections, only a fraction of which are histologically examined.176 On the other hand, the presence of metastasis in an excised node can be determined by PAI257 since most melanomas are highly melanotic, containing 95% melanin,11 and melanin is a highly absorbing chromophore in NIR range.115 Therefore, SLN metastases can be accurately identified by PAI, leading to appropriate selection of sections for histological examination, mitigating the possibility of false negative results.258

3.3.1.

Animal studies

PAI has been studied for the imaging of lymph nodes (Table 3) to identify metastases both ex vivo (postsurgical resection) and in vivo. McCormack et al.176 used a three-single-element-transducer based PAT prior to and after injecting a human melanoma cell line (HS 936) into ex vivo lymph nodes from a healthy canine and pig. The PAT system they used consisted of a 600  μm diameter optical fiber connected to a tunable laser and an acoustic sensor made from polyvinylidene fluoride film. The 532 nm wavelength light was illuminated from the top of the lymph node, and the PA signals were detected from the bottom [see Fig. 7(a)]. The control lymph nodes (with no injected melanoma cells) showed no PA response [see Fig. 7(a)(i)], whereas the melanoma cells in the excised lymph nodes generated PA signals as shown with the arrow in Fig. 7(a)(ii). A pig lymph node with only 500 injected melanoma cells and a 100 to 200  μm diameter lymph node produced a PA response. Neuschmelting et al.180 utilized a multispectral (wavelengths 700 to 860 sampled) PAT system with a cylindrically focused transducer array (iThera Medical) and compared it with fluorodexyglucose (FDG) positron emission tomography (PET)/CT for imaging B16F10-derived melanoma micrometastases and macrometastases to lymph nodes and in-transit metastases (metastases moving from the primary tumor to the nearby nodal basin) in mice in vivo. The PAT system detected lymph node micrometastases in the cortical region of lymph nodes (after 2 weeks’ tumor cell inoculation) that were too small for FDG PET/CT to detect. It also delineated in-transit metastases, which were observed as bright clusters between the primary melanoma site and the nodal basin. Both PAT and PET/CT could detect macrometastases [see Fig. 7(b)], but only the PAT system unmixed signals to enable detection of micrometastatic infiltration of melanoma in the cortex of popliteal nodes (white dashed circles). This multispectral PAT system distinguished melanoma lymph node metastases from other neoplastic and nonneoplastic lymphadenopathies (due to melanin’s contrast) but FDG PET/CT could not (due to nonspecific FDG uptake and relatively low resolution). Most recently, Sinnamon et al. utilized a Vevo LAZR PAT system to visualize inguinal lymph node metastases in vivo at 4 and 8 weeks after inoculation with B16 melanoma cells in the flank region in BRaf-PTEN transgenic mice.174,183 In total, 49 lymph nodes were imaged in 25 mice, with metastatic cells present in 17 lymph nodes (35%), with histopathological confirmation. Thus the system was able to image in vivo melanin PA signals of a lymph node containing melanoma metastasis at 60 days after the induction of the tumor [see Fig. 7(c)(i)]. The negative control mouse without tumor induction is shown in Fig. 7(c)(ii). The PA melanin signal within the positive lymph nodes was significantly higher than in the negative control mouse, with no difference in PA signals between the adjacent soft tissue of positive and negative lymph nodes. The strongest predictor of melanoma metastasis was the ratio of lymph node to soft tissue PA melanin peak signal.

Table 3

Summary of ex vivo and in vivo human and animal PAI studies for detection of melanoma metastasis to lymph nodes.

PAI modalityLight sourceUS transducerImaging modelStudy result
LA-PATλ: 680 to 970 nmCF: 21 MHzHuman lymph nodes imaged both ex vivo and in vivo174Identification of melanoma metastasis in human lymph nodes
BW: 70%
NE: 256
LA-PAT/multispectralλ: 700 to 860 nmCF: 5 MHzMouse lymph nodes imaged in vivo180Detection of lymph node micro metastases and in-transit metastases from melanoma
BW: NL
NE: 256
Vevo LAZRλ: 680 to 970 nmLZ250Mouse lymph nodes imaged in vivo183Melanin detection in melanoma metastases. The strongest predictor was the ratio of lymph node to soft tissue PA melanin peak signal
CF: 21 MHz
BW: 55%
NE: 256
LZ201
CF: 15 MHz
BW: 60%
NE: 256
PATλ: 532 nm2.5  μm thick PVDF film and coaxial cablePig and canine lymph nodes imaged ex vivo176Detection of melanoma micro metastases in SLNs
PATλ: 720 to 800 nmCF: 6.25 MHzHuman lymph nodes imaged ex vivo169First human study on melanoma lymph node metastasis. Examining the status of resected lymph nodes
BW: >80%
NE: 32
Photoacoustic finderλ: 532 nmCF: 8 MHzMouse lymph nodes imaged in vivo239Use of PA for localization of the SLNs in rats without exogenous agent
λ: 650 nmCW: 45%
NE: 1
PATEx vivo:CF: 5 MHzHuman lymph nodes imaged ex vivo from patients with stage I and II melanoma117Imaging of SLNs to determine metastatic status
λ: 700–880 nmBW: 60%100% sensitivity and 62% specificity
NE: 128
PATIn vivo:2D:Human lymph nodes imaged in vivo from patients with stage I and II melanoma117ICG dye used to mark SLNs. 100% sensitivity and 48.6% specificity
λ: 700, 730, 760, 800, 850 nmCF: 4 MHz
BW: 52%
NE: 256
3D:
CF: 2.5 MHz
BW: 60%
NE: 384
LA-PATλ: 532, 650, 1064 nmCF: 5.3 MHzRat lymph nodes imaged in vivo141The SLN was detected in the rat beneath 2.2 cm thick chicken tissue layer, at a depth that is at least typical of SLN in humans
BW: 90%
NE: 192
Note: λ, wavelength; F, fluence; CF, central frequency; BW, bandwidth; NE, number of elements; NL, not listed; LA-PAT, linear array photoacoustic tomography; PAFC, photoacoustic flow cytometry; PAF, photoacoustic finder; AR-PAM, acoustic resolution photoacoustic microscopy; OR-PAM, optical resolution photoacoustic microscopy; SLN, sentinel lymph node.

Fig. 7

Animal studies of lymph node metastases analyzed by PAI. (a) Photoacoustic response from lymph nodes (i) without melanoma, the detector shows no signal with a noise floor of 100  μV, and (ii) with melanoma, showing a photoacoustic wave at 5  μs. Reproduced with permission from Ref. 176. (b) Comparative results between multispectral PAT and FDG-PET/CT systems for detection of macrometastasis in right popliteal lymph node in mice. The red circle encloses the melanoma macrometastasis region. White circle enclosed the contralateral popliteal healthy control node. Multispectral images were unmixed for deoxyhemoglobin (blue), oxyhemoglobin (red) and melanin (yellow). Fb, femur; Tv, tail vessel; Rc, rectum; Ut, urethra. Reproduced with permission from Ref. 180. (c) In vivo images of melanin-specific photoacoustic signal with grayscale underlay of a lymph node that (i) shows metastasis at 60 days post tumor induction and (ii) negative control mouse that did not undergo tumor induction. MPAI, multispectral photoacoustic imaging; FDG-PET/CT, fluorodexyglucose PET/CT. Reproduced with permission from Ref. 183.

JBO_29_S1_S11518_f007.png

3.3.2.

Human studies

Many different PAI modalities have been utilized for ex vivo studies of metastases in melanoma SLNs, including LA-PAT and arc-shaped PAT.117,169,174 PAT has also been used for in vivo studies.117 The first human study on melanoma lymph node metastasis was conducted by Grootendorst et al.169 using a curvilinear array. Patients with proven metastatic disease undergoing inguinal or axillary lymphadenectomy were enrolled, and 1 or 2 lymph nodes were randomly selected for ex vivo multispectral analysis. The PA laser was illuminated from the top of the sample and the US detector array was rotated 360 deg around the sample. A total of six lymph nodes were imaged, and PAT revealed that three lymph nodes were metastatic and three were benign. All three malignant nodes (LN1, LN2, and LN3) displayed increased PA signals (from melanoma cells), whereas all three benign nodes had a substantially weaker signal (signal likely from hemoglobin and possibly other chromophores) when imaged at wavelengths from 720 to 800 nm [see Fig. 8(a)]. The results were confirmed through histopathology. Langhout et al.174 used a LA-PAT system and imaged 12 lymph nodes. Histopathology revealed that three nodes were metastatic and nine were benign. Again, melanoma-positive nodes [see Fig. 8(b)(i)] displayed different PA signals (depth image) than benign nodes [see Fig. 8(b)(ii)] due to the difference in melanin distribution. Additionally, total volume imaging to the depth of 2 cm in benign lymph nodes (absent melanin deposits) allowed for computation of the entire nodal volume. The largest human study on PAI of melanoma metastasis to lymph nodes was carried out by Stoffels et al.117 using a multispectral arc-shaped PAT. They analyzed 506 SLNs from 214 patients with a Breslow depth of at least 1 mm: 148 SLNs (from 65 patients) were analyzed by multispectral PAT ex vivo and histology, whereas the other 358 SLNs (from 149 patients) were analyzed by the conventional European Organization for Research and Treatment of Cancer (EORTC) melanoma group protocol. Their system detected metastases in 22.9% of excised SLNs compared with 14.2% by the EORTC Melanoma Group protocol. Ex vivo analysis by PAT showed 100% sensitivity and 62% specificity. Then, an in vivo experiment was carried out using PAT and indocyanine green (ICG) (a NIR fluorophore injected peri-tumorally) as a contrast agent to image 41 SLNs in 20 patients. PAT visualized ICG-marked SLNs to the depth of 5 cm (discerned by single photon emission computed tomography/CT) and with 100% concordance with the gold standard of SLN detection, 99 m Tc-nanocolloid–guided lymphoscintigraphy. In vivo PAT analysis revealed a sensitivity of 100% and specificity of 48.6%. With 100% sensitivity, both ex vivo and in vivo PAT identified noncancerous SLNs in 189 total lymph nodes without any false negatives but with a high rate of false positives. The quantification of melanin was performed with multispectral analysis and then correlated with the localization of metastatic cells [see Fig. 8(c)].

Fig. 8

Ex vivo and in vivo studies of human lymph node metastases analyzed by PAI. (a) PA signal strength of the selected areas within the lymph nodes (LN) at different illumination wavelengths. Reproduced with permission from Ref. 169. (b) Images of two human nodes. (i) PA image of a metastatic node, (ii) PA image of a benign node. Absence of PA signal deeper in the malignant node [as indicated by * in panel (b)] seems to be caused by the strong absorption by the melanin in the superficial area of the node. Reproduced with permission from Ref. 174. (c) (i) Lateral MIP and (ii) 3D rendering image of ex vivo optoacoustic image of a human lymph node from a melanoma patient. Grayscale represents hemoglobin background and color bar overlay shows the multispectral resolved signals for melanin. Reproduced with permission from Ref. 117.

JBO_29_S1_S11518_f008.png

3.4.

Metastases to the Blood: Imaging CTCs

CTCs originate in the primary tumor and acquire genetic and structural alterations, leading to changes in cellular signaling and protein expression. This results in bloodstream intravasation and possibly invasion to other organs to develop secondary tumors.259,260 Previous work has shown that CTC assays can predict prognosis and treatment response in patients with breast, prostate, and colorectal cancers.261263 CTCs can be detected via flow cytometry, which has traditionally involved detecting fluorescent signals from cells labeled with multicolor probes ex vivo, which is known as conventional flow cytometry.264,265 However, this method requires invasive blood extraction from patients and is limited in its ability to detect infrequent CTCs.265 Additional CTC assays have been developed,266 but have not been studied on a large scale, limiting their clinical translatability.

Flow cytometry can be done in vivo and noninvasively using PA flow cytometry (PAFC). The concept of PAFC is similar to PAI: an excitation light is absorbed by chromophores in blood cells, causing local thermoelastic expansion and subsequent production of acoustic waves, which can be detected by an US transducer. PAFC configurations are an adaptation of OR-PAM, as high-pulse-repetition-rate lasers are also used in PAFC to generate detectable PA signals from individual CTCs.265 Higher pulse rates improve the signal-to-noise ratio (SNR) through averaging PA signals from individual CTCs; SNR is determined by the ratio of PA signals from individual CTCs to signals from other blood cells and background noise.265 The main chromophore in blood cells is hemoglobin in red blood cells (RBCs), but the main chromophore in melanoma CTCs is melanin. As with many other PAI modalities, in PAFC, multispectral analysis is often used to differentiate hemoglobin/melanin signals in the vasculature. Thus, unmixing of multispectral PA signals can allow for differentiation of CTC signals from oxy- and deoxy-hemoglobin.167 Numerous studies, summarized in Table 4, have investigated PAFC to detect melanoma CTCs.137,139,166168,170172,178,179,189,190

Table 4

Summary of ex vivo and in vivo human and animal PAI studies on melanoma CTCs.

PAI modalityLight sourceUS transducerResolutionImaging modelStudy result
PAFCλ: 420 to 2300 nmCF: 10 MHzNLIn vivo: Imaging of the carotid artery of mice168Develop in vivo lymph tests using the principles of flow cytometry. Calculated the CTC flow rate
BW: NL
NE: 1
PAFCλ: 905 nmCF: 3.5 MHzLR: 100  μmIn vivo: Mice tail veinDetection of CTCs. About 82% of unlabeled CTC were detected
CF: 20 MHzAR: 15  μm
BW: NLLabel: AuNR
NE: 1Mice ears and skin. No label167
PAFCλ: 430 to 630 nmPVDF transducerNLIn vitro melan-oma cells tagged with AuNP178The AuNP tagged melanoma cells showed 34% greater PA signal than the untagged cells
BW: NL
NE: 1
PAFCλ: 820 to 1064 nmCF: 3.5 MHzNLIn vivo: Mice tail vein, ears vessels, and abdominal vessels190Detection of CTCs in melanoma-bearing mice. PA signals were detected immediately after injection. Calculation of the CTC flow rate
BW: NL
NE: 1
PAFCλ: 1060 nmPVDF transducerLR:In vitro: Detection of CTCs spiked in blood samples. In vivo: Assessment of effect of optical clearing on mice and humans179Detection of melanoma cells in blood flow
40×1100  μm
CF: 16 MHzAR: 20  μmOptical clearing increased PA signal amplitude by 20% to 40%
BW: 190%
NE: 1
PAFCλ: 532 nmCF: 10 MHzLR: 1.5  μmIn vivo: Mice, subcutaneous injection, detection of CTCs in ear189Identification and count of RBCs and CTCs in the blood vessels
BW: NLAR: 132  μm
NE: 1
PAFCλ1: 532 nmCF: 50 MHzLR: 3 to 7  μmIn vivo: Mice, injection of B16 cells in jugular vein or carotid artery172Detection of CTCs and clusters traveling in blood vessels
λ2: 1064 nmBW: NLAR: 26  μm
NE: 1
PAFCλ: 1060 nmPVDFLR: 65  μmIn vivo: Healthy humans and humans with melanoma166Detection of melanin-bearing CTCs in patients with melanoma. CTC flow rate was calculated. PAFC distinguished CTCs from circulating blood clots
CF: 16 MHz
BW: 190%
NE: 1
PAFCλ1: 532 nmPVDF transducerNLIn vitro: Humans with melanoma170Detection of CTCs. Signal from a single melanoma cell is detectable
λ2: 680 nm
CF: NL
BW: NL
NE: 1
PAFCλ: 532 nmPVDF transducerNLIn vitro: Blood samples with cultured melanoma cells207Detection of melanoma CTCs in blood samples
CF: 50 MHz
BW: 100%
NE: 1
PAFCλ: 532 nmNLNLIn vitro: Blood samples of patients137Detection of melanoma in stage IV patients
PAFCλ: 1064 nmCF: 20 MHzNLIn vitro and in vivo: Mice blood or mice with injected B16 cells213Detection of melanoma with nonlinear PAFC
BW: NL
NE: 1
PAFCλ1: 532 nmNLLR: 30  μmIn vivo: CTCs in mice, rat tail artery208PA detection of induced metastatic melanoma
λ2: 622 nm
PAFCλ: 820 nmCF: 10 MHzNLIn vivo: Mice artery after tail injection211Detection of strongly absorbing cells with positive contrast and negative contrast for fluorescence
NE: 1
PAFCλ: 750 nmCF: 40 MHzLR: 40  μmGelatin phantom with solution containing CTCs214Monitoring of low concentration of CTCs without labeling
BW: 85%
NE: NL
PAFCλ1: 670 nmCF1: 3.5NLIn vivo and ex vivo: Mice brain212,215Detection and counting of CTCs in CSF of tumor-bearing mice
λ2: 820 nmMHz
CF2: 20 MHz
PAFCλ: 1064 nmCF: 3.5 MHzNLIn vitro and in vivo: Melanoma cells in mouse blood and in mice paw138Reliable detection of melanoma cells in the vessels after upstream arterial injection
NE: 1
PAFCλ: 820 nmCF: 2.25 MHzNLIn vivo: Mice ears193Detection of lymphatic CTCs in premetastatic disease
NE: 1
PAFCλ: 532 nmCF: 5 MHzNLIn vitro: Human blood samples with stage I, II, and III melanomas199Quantification of CTCs in serial blood samples at early stage melanoma to predict metastatic disease
NE: 1
OR-PAFCλ: 1064 nmCF: 40 MHzLR: 15  μmIn vitro: Bovine blood samples with melanoma cells140Detection of melanoma cells flowing in blood sample. Estimation of the flow speed of the cell in the fluid
BW: 100%AR: 37  μm
NE: 1
LA-PATλ: 680 nmCF: 21 MHzLR: 119  μmIn vivo: Mice tail veins171Detection and quantification of CTCs
BW: 78%AR: 86  μm
NE: 256
LA-PATλ: 680 nmCF: 40 MHzLR: 94  μmIn vivo: Humans with melanoma192Imaging suspected CTCs in patients in vivo, with a CNR >9
BW: 85%AR: 43  μm
NE: 256
SIP-PATλ: 680 nmCF: 5 MHzSR: 125  μmIn vivo: Mice carotid artery139Visualization of CTCs in mice cortical arteries and veins and calculation of the CTC flow rate
BW: 90%
NE: 512
PATλ: 700 nmCF: 5 MHzSP: 150  μmIn vivo: Mice brain with melanoma CTCs191Detection and counting of individual CTCs in the CSF
BW: 100%
NE: 512
Note: RBC, red blood cell; λ, wavelength; CF, central frequency; BF, broadband frequency; BW, bandwidth; NE, number of elements; NL, not listed; AuNR, gold nanorods; LR, lateral resolution; AR, axial resolution; AR-PAFC, acoustic resolution PA flow cytometry; OR-PAFC, optical resolution PA flow cytometry; SIP-PAT, single pulse panoramic PAT; PVDF, polyvinyledene fluoride; CNR, contrast-to-noise ratio; CSF, cerebral spinal fluid.

3.4.1.

Animal studies

PAFC has demonstrated the ability to detect unlabeled (and labeled) CTCs in the mouse vasculature after injection of CTCs in the tail vein or from inoculation of primary melanoma tumors in the skin or ears.87,167,168,172,189,190 Overall, PAFC accurately detected CTCs from the background RBCs in the mouse vasculature, and PAFC signals increased as metastasis increased.139,167,168,171,172,189,190 Deán-Ben et al.191 employed spherical array PAT for real-time visualization of passage and trapping of individual B16 melanoma cells in the whole mouse brain. Imaging was performed with the laser wavelength and pulse repetition rate set to 700 nm and 50 Hz, respectively. About 100 frames of PA images were acquired before injection of B16 melanoma cells and averaged to determine a baseline. The system could identify and track B16 melanoma cells after injection by taking the difference between before and after images.191

3.4.2.

Human studies

Galanzha et al.166 studied CTCs in healthy patients and patients with melanoma using PAFC in vivo. PAFC data from healthy patients were used to calculate false positives and to study PA artifacts.166 PAFC accurately identified unlabeled CTCs in 15/16 melanoma patients.166 The system uses MPAI with one channel for detection of blood and a second channel for melanin. The channels are shown individually and fused together in Fig. 9(a)(i), and fused images showing movement of melanoma CTCs in an artery [Fig. 9(a)(ii)] and a vein [Fig. 9(a)(iii)] are also presented. This system has relatively poor lateral resolution (Table 4), which the authors explain is caused by light beam blurring.166 Recently, PAT systems have also been used to detect CTCs. Hai et al.171,192 employed a LA-PAT system to detect melanoma CTCs in patients in vivo. Based on the optical absorption coefficient ratio, an excitation wavelength of 680 nm was chosen to maximize the contrast between melanoma CTCs and blood and achieve the highest detection sensitivity. Contrast-to-noise ratio (CNR) was used to quantify melanoma CTCs from the background tissue (RBCs). From the imaging session of the forearm of a positive patient with stage IV metastatic melanoma, a CTC was detected with a CNR of 9.4 [see Fig. 9(b)]. The single cells were captured at five frames and the flow speed was estimated to be 9.6  mm/s. They imagined 16 stage III and IV melanoma patients and successfully detected suspected melanoma CTCs in three patients. In fact, two of the three had disease progression, but four of those found CTC-negative also had disease progression. The lower rate of CTC detection by LA-PAT compared with PAFC could be attributed to the use of melanin as the single marker to identify and detect CTCs, and the fact that the patients were measured at only one clinical time point instead of on multiple repeat visits.171,192 Also, in the previous study, optical clearing techniques, including microdermabrasion and glycerol sonophoresis, increased PA signals by two- to threefold, due to the reduced light scattering in superficial skin layers.166 PAT has tremendous clinical potential in imaging CTCs and disease monitoring in melanoma, given the fact that PAT can image very deep in the tissue with very high optical contrast. Moreover, utilization of contrast agents for molecular PAI can further increase the sensitivity of PAT to imaging cancer cells that do not express melanin.

Fig. 9

Imaging melanoma circulating tumor cells in vivo. (a) PA flow cytometry: channels shown individually and fused together (i), fused images showing movement of melanoma CTCs in an artery over time (ii), and in a vein (iii). (b) LA-PAT system uses differential analysis of images taken over time to find moving CTC cells: (i) PA snapshots of the melanoma CTC in the patient. The yellow arrows indicate structures, including the skin, vessel boundaries, and subcutaneous fat layer. The red arrows highlight the melanoma CTC. (ii) Differential PA images showing only the melanoma CTC. (iii) Differential PA images superimposed on structural images, highlighting the melanoma CTCs. Reproduced with permission from Ref. 192.

JBO_29_S1_S11518_f009.png

3.5.

Virtual Histology

Recently, all-optical PA microscopy in reflection-mode has been shown to be able to form histology-like images of various cancers on unstained slides to distinguish tissue types.142,267 The principle behind this technology is that by concurrently measuring radiative and nonradiative (in the form of acoustic) emissions from tissue samples illuminated with light in the UV range (266 nm), it is possible to differentiate DNA, RNA, collagen, and elastin, among other chromophores. Nuclear contrast, in particular, comes from relaxation of DNA, and nonnuclear contrast from relation of extranuclear proteins. These enable high-resolution images remarkably analogous to traditional chemical haematoxylin and eosin (H&E) staining.267,268 This technique has very recently been applied to the analysis of skin biopsies suspected for melanoma.269,270

3.6.

Exogenous Contrast Agents for Melanoma Detection

PAI most commonly utilizes intrinsic (endogenous) contrast agents, such as melanin and oxy- and deoxy hemoglobin to assess melanomas.271 However, an exogenous, melanoma-specific contrast agent could assist in assessing early-stage melanomas, tumor staging, surgical excision planning, detection of lymph node metastases, and detection of CTCs. In Table 5, some of these applications are demonstrated. Gold nanoparticles (AuNPs), due to their intrinsic bioinertness and highly tunable optical properties, have shown remarkable promise in cancer diagnostics and treatment.273 As a contrast agent, AuNPs display optical absorption and scattering cross-sections that are greater than those of organic dyes, making them appropriate contrast agents for several applications in biomedical optics, including PAI.

Table 5

Use of exogenous PA contrast agents for melanoma detection in mice.

PAI modalityLight sourceUS transducerResolutionAgentImaging modelStudy result
AR-PAMλ1: 778 nmCF1: 50 MHzLR: 45 to 506  μmAuNCMice on the dorsal surface173Volumetric images illustrate both tumors and blood vessels
λ2: 570 nmCF2: 10 MHzAR: 15 to 150  μm
NE: 1
LA-PATλ: 780 nmCF: 21 MHzAR: 75  μmMAGE-Au-PFH-NPsMice, subcutaneous175Tumor detection from enhanced PAI signal
BW: 70%
NE: 256
OR-PAMλ1: 639 nmCF: 3.5 MHzNLMNPs/Evans Blue dyeMouse lymph nodes imaged in vivo165Detection and treatment of metastases in SLN at a single cell level. Detection of micro-metastasis
λ2: 850 nmBW: NL
NE: 1
PAFCλ: 808 nmCF: NLNLID-HCuSNP, DOX and ICGMice, tail vein195Cell membrane-camouflaged NPs have excellent self-recognition ability to the aimed tumor cells in vivo
BW: NL
PAFCλ: 420 to 2300 nmCF: 10 MHzSR: 6 to 20  μmAuNRsMice, tail vein272Better absorption contrast introduced by AuNRs than control melanoma cells
BW: NL
NE: 1
PAFCλ: 860 nmCF: NLNLAuNPs in colony forming cells (ECFCs)Mice, Tumor-bearing196AuNP-loaded ECFCs generate higher PA signals than AuNPs alone
BW: NL
Note: λ, wavelength; CF, central frequency; NE, number of elements; BW, bandwidth; LR, lateral resolution; AR, axial resolution; SR, spatial resolution; NL, not listed; PAI, photoacoustic imaging; AR-PAM, acoustic resolution photoacoustic microscopy; LA-PAT, linear array photoacoustic tomography; AuNC, gold nanocrystals; MAGE-Au-PFH-NPs, Au nanorods and liquid perfluorocarbon (PFH) conjugated to a MAGE-1 antibody; ID-HCuSNP, hollow copper sulfide nanoparticle; DOX, doxorubicin; ICG, indocyanine green; PAFC, photoacoustic flow cytometry.

For example, gold nanorods (AuNRs) have been used to increase optical contrast in CTCs.272,274 Zharov et al.272 used PAFC to detect CTCs (human squamous carcinoma cell line SQ20B) labeled with AuNRs, which were 15  nm×52  nm in size and had a maximum absorption at 840 nm. The cancer cells labeled with AuNRs were injected into the mouse circulatory system through the tail vein. The real-time accumulation of AuNRs in the cells was monitored through an increase in PA signal. The maximum absorption contrast introduced by AuNRs compared with background tissue was 291. The capability of AuNR was compared with that of a conventional contrast agent called ICG. At the same laser energy, linear PA signals from AuNR-labeled cancer cells were five to seven times stronger than the signals from cancer cells stained with ICG.

3.6.1.

Animal studies

Kim et al.173 studied PA skin imaging of gold nanocrystals (AuNCs) bio-conjugated to [Nle4,D-Phe7]-α-melanocyte-stimulating hormone ([Nle4,D-Phe7]-α-MSH) and compared them to poly(ethylene glycol) (PEG)-AuNCs. Melanoma cells strongly overexpress α-MSH receptors. In their study, mice were inoculated with B16 melanoma cells and either one or the other contrast agent was injected via tail vein. In vitro studies showed that melanoma cellular uptake of [Nle4,D-Phe7]-α-MSH-AuNCs was 3.5 times greater than PEG-AuNCs uptake after 6- and 24-h incubation periods. In vivo experiments involved two groups of mice: one group (n=4) received [Nle4,D-Phe7]-α-MSH-AuNCs and the other group (n=4) received PEG-AuNCs, both via tail vein [see Fig. 10(a)]. Six hours after injection, the PA signals of [Nle4,D-Phe7]-α-MSH AuNCs increased 38%, whereas the PEG-AuNCs increased only 13% [see Fig. 10(a)(iv)]. The number of AuNCs was quantified in the excised tumors using inductively coupled plasma mass spectrometry. The mean number of [Nle4,D-Phe7]-α-MSH AuNCs per gram of tumor was found to be 360% times greater than that of PEG-AuNCs, indicating the extremely high uptake of [Nle4,D-Phe7]-α-MSH by melanoma cells. More recently, Li et al.175 evaluated a nanoparticle composed of AuNRs and liquid perfluorocarbon (PFH) and conjugated it to a monoclonal antibody to melanoma-associated antigens (MAGE-1 antibody) and used an LA-PAT system for imaging. In vitro experiments showed a high number of MAGE-Au-PFH-NPs concentrated in the plasma membrane and cytoplasm of melanoma cells, compared with Au-PFH-NPs (control), which only showed a weak binding around tumor cells. Laser irradiation of nanoparticles with PFH results in a phase change (liquid PFH to gas microbubbles), increasing the acoustic impedance of surrounding tissues and subsequently causing a signal enhancement. In vivo PAI in tumor-bearing mice revealed a significantly higher PA signal intensity 2 h after MAGE-Au-PFH-NPs injection into the tail vein compared to Au-PFH-NPs; higher conjugate concentrations produced greater PA signals [see Fig. 10(b)].

Fig. 10

In vivo animal studies involving exogenous PAI contrast agents for melanoma detection. (a) In vivo MAP images of B16 melanomas using [Nle4,D-Phe7]-α-MSH- and PEG-AuNCs. (i), (ii) Photographs of tumor in mice before injection. (iii), (v), and (vii) Time course PA images after injection of [Nle4,D-Phe7]-α-MSH-AuNCs. (iv) (vi), and (viii) Time course PA images after injection of PEG-AuNCs. Reproduced with permission from Ref. 173. (b) PA images of B16 melanomas in mice at different time points after the injection of (i) MAGE-Au-PFH-NPs and (ii) AU-PFH-NPs. Reproduced with permission from Ref. 175. (c) PA imaging of melanoma in mice (i) before and (ii) after the injection of ID-HCuSNP@B16F10 NPs. Reproduced from Ref. 195. MAP, maximum amplitude projection.

JBO_29_S1_S11518_f010.png

Wu et al.195 developed cell membrane-camouflaging hollow copper sulfide nanoparticles (ID-HCuSNP) by coating the membrane of melanoma cells with doxorubicin and ICG-loaded hollow copper sulfide NPs to enhance their targeting ability. They injected a mouse model with ID-HCuSNP and after 4 h, and they observed a strong local PA signal in the tumor area due to the accumulated NPs.195 This showed cancer cell membrane-camouflaged NPs having an excellent self-recognition ability to the aimed tumor cells in vivo [see Fig. 10(c)]. Galanzha et al.165 used PAT with mapping techniques (multicolor PA lymph flow cytometry, PA lymphography, and absorption image cytometry) to examine metastasis to the prenodal lymph vessels and SLNs in mice ex vivo and in vivo. Specifically, after a visible primary melanoma tumor had developed in the mouse ear, they injected magnetic nanoparticles (MNPs) that provided stronger PA signals from lymphatics and SLNs at 639 nm. However, similar NIR absorption spectra of MNPs and melanin made it difficult for spectral identification of melanoma metastasis in the presence of MNPs. To resolve this problem, they used Evans Blue (EB) dye, which absorbs poorly at 639 nm only, while the presence of melanoma cells in lymphatics and in a node was identified through the appearance of PA signals at 850 nm. Because absorption of melanin also occurred at 639 nm above the background from EB dye, they used specific ratio of PA signals at 639 and 850 nm for additional identification of melanoma cells. They injected EB dye near melanomas derived from B16F10 cells and imaged the region in week 1 and 2 postinoculation. The number of metastatic melanoma cells in transit increased from 0.26  cells/min in week 1 postinoculation to 2.13  cells/min in week 2 postinoculation. The percentage of PA signal covering the examined SLN area increased from 6% in week 1 postinoculation to 39% in week 2 postinoculation. In the histology results, lymph nodes showed no metastases after 1-week inoculation and single metastases after 2 weeks inoculation. Thus the PA system was able to detect early micrometastases that could not be visualized in histology, illustrating its ability to detect even single metastatic cells (at week 1 postinoculation).

Despite the number of reports on the successful demonstration of Au nanomaterials for cancer theranostics,275,276 accumulations in the liver and spleen due to resident macrophages that form the mononuclear phagocyte system are still an issue.277,278 Armanetti et al.196 utilized endothelial colony forming cells (ECFCs) to carry AuNPs and explored the antitumor effects and the tumor-homing efficiency following single intravenous injection into tumor-bearing mice. They assessed AuNPs biodistribution in freshly excised mice organs at different time points post administration by exploiting the PA properties of AuNP-enriched ECFCs. They demonstrated in vitro that AuNP-loaded ECFCs are able to generate higher PA signals than AuNPs alone and also display spectral fingerprints that enable a reliable detection of labeled cells following intravenous injection.196

Toxicity is always a concern when developing exogenous contrast agents. Factors for assessing biocompatibility include inertness, metabolism, and effective clearance rates. Material composition, surface modifications, shape, and size must all be designed carefully to maximize biocompatibility. The growing field of PAI for cancer diagnosis and treatment planning is accelerating research in biocompatible exogenous contrast agents.279,280

4.

Discussion

The scalability of PAI technology (from OR-PAM to AR-PAM to PAT) allows a wide range of applications including melanoma detection and depth determination, tumor-related angiogenesis, lymph node metastases, and presence of CTCs, all either with endogenous or exogenous contrast agents. Although these applications have been explored using small animal models for a couple dozen years, in the past decade or so, human observational studies have demonstrated the clinical potential for these systems. Although these technologies need more confirmational human studies, they lay a strong foundation for the near-term translation of PAI systems to the clinic for melanoma disease detection and management.

In terms of initial diagnosis and melanoma depth determination, if melanoma is in its very earliest stages (at the epidermis), OR-PAM provides rich information on tumor presence and angiogenesis—therefore, it can be used not only for detection but for accurate treatment planning, as the extent of angiogenesis may predict tumor aggressiveness. AR-PAM could be a better choice for deeper melanomas (approximately 1  mm). Although PAT could be implemented using ultrahigh frequency transducers, the use of it to detect and characterize early stage melanomas has not been explored, perhaps due to the requirement for a much-more sophisticated data acquisition system, transducer focal length, and complex light illumination configuration. Another shortcoming of PAT is that it is limited to providing 2D images, unless it is implemented with a scanning system, which adds complexity. Nevertheless, tunable lasers with a wide range of wavelengths (to accurately distinguish melanoma biomarkers such as melanin) are much more accessible for PAT implementation compared to AR/OR-PAM. Therefore, where permitted, high or ultrahigh frequency PAT could be a good choice for melanoma detection and depth determination. Complementing current histopathology of excised tissue, recently, all-optical PA microscopy in reflection-mode has been shown to form histology-like images of various cancers on unstained slides to distinguish tissue types. This technique has great potential to be in the clinic for rapid dermatological tissue analysis. Once a melanoma is appropriately detected, knowing whether it has metastasized to the SLN is the next important question for treatment planning. Because of the depth of SLNs, PAT might be the best choice, and in fact, in Table 3, all of the human studies of SLNs were performed by PAT, although in some cases, AR-PAM could potentially be used as well. In cases where melanoma is confirmed metastasized to the SLN (or beyond), detection of CTCs is very valuable for monitoring responses to cancer treatment and evaluating prognosis. With PAI being a technology based on high-energy laser, fast switching between imaging and ablation of CTCs has been investigated in small animals, it could be translated into a theranostic application, once the concept is sufficiently confirmed through clinical studies.

Although melanin is an adequate optical contrast for early-stage melanomas, it is not sufficient for later stage melanomas due to the limitation of light penetration in deep structures. Exogenous contrast agents have been shown to increase PA signals beyond the signal level obtained using the label-free PAI (endogenous contrast agents). As discussed in Sec. 3.5, different tumor cell targets (antigens) of the antibody-coupled exogenous contrast agents have been tested on animal models. While the results seemed promising, exogenous contrast agents need to be proven safe to have clinical utility. In addition, before exogenous contrast agents are studied in humans, a unique target protein present on melanoma cells and absent on healthy cells will need to be identified, such as galectin-3 or collagen XVII, which have recently been shown to be overexpressed in melanoma cells.281 Furthermore, few if any studies have investigated whether a contrast agent could be applied topically on the melanoma lesion as opposed to intravenously, which could mitigate potential toxicity. However, PAI will likely need to gain clinical utility before exogenous contrast agents are rigorously investigated in humans.

While PAI holds great clinical potential, safety and cost are factors that may limit the ease of clinical translatability. There is a safety concern about tissue damage from the light excitation. The American National Standards Institute has already defined the maximum permissible exposure, which is the level of electromagnetic radiation that a person can be exposed to without harmful effects, for skin imaging;91 another is potential damage to the eyes of the patient or the physician from stray laser light, which can be mitigated by using appropriate protective eyewear. Cost is a further potential limitation that can be mitigated by mass production of PAI systems. A final hurdle is the need for pulsed nanosecond lasers, which can be costly. Low-cost laser sources such as laser diodes and light-emitting diodes (LEDs) have been shown to be effective in some PAI applications.282284

In recent years, there has been a growing interest among various companies in the development and commercialization of PAI systems for medical applications.145,285 Among them are iThera Medical GmbH,105,130,180,286297 FUJIFILM VisualSonics,135,171,187,298 ENDRA Life Sciences Inc.,299303 TomoWave Laboratories,304307 and Seno Medical Instruments.308,309 Although there exist commercial systems for skin imaging, there has not been any customized clinical system for melanoma imaging applications.

5.

Conclusions

Prior CM studies in animals and humans have illustrated PAI’s potential use in identifying tumor depth, analysis of angiogenesis, detection of lymph node metastasis, detection and ablation of CTCs, and ability to generate histology-like images of various cancers on unstained slides. These applications stem from the high spatial and temporal resolution, adequate penetration depth, and label-free nature of this imaging modality.

Many of these opportunities are based on the use of melanin as a high-absorbing chromophore. However, reliance on melanin has a few limitations. First, as noted above,251 for tumors with very high density of melanoma, it can be difficult for PAI to penetrate through the tumor to detect lower boundaries without assistance from, for example, US structural information. Second, amelanotic melanoma, which accounts for 1% to 8% of all melanomas, is a form of melanoma with little to no pigment (lesion pigment not differentiated from skin).310 Methods focused on melanin detection could miss these lesions, and PAFC likely will miss amelanotic CTCs.

Despite the advantages of PAI described in this review, this technique has not yet been used as a diagnostic-assistant tool in large-scale clinical studies. One possible way to enroll large groups of patients in future studies would be to incorporate PAI before performing procedures to remove lesions suspicious for melanoma or before lymph node excisions. This would allow for characterization of PAI images from various pigmented lesions that may clinically resemble melanoma, such as benign nevi, dysplastic nevi, lentigo, etc., as well potential malignant lymph nodes. As more images are collected and analyzed, algorithms on the recognition of melanoma in PA images will mature.

Safety and cost are two key parameters in clinical translatability of PAI. The use of laser light must be limited to the maximum permissible levels for skin imaging, and protective eyewear or confining apparatus can mitigate potential damage to eyes. PAI system costs can be reduced by using a combination of LEDs and development of low-cost US transducers designed specifically for PA applications.

Disclosures

The authors declare no conflict of interest.

Code and Data Availability

The data presented in this review article has been collected from different sources and each source has been cited in the article for further details.

Author Contributions

Joseph Fakhoury contributed to investigation, visualization, and writing—original draft, review, and editing. Juliana Benavides contributed to investigation and writing—review and editing. Rayyan Manwar contributed to visualization, and writing—original draft, review, and editing. Mohsin Zafar contributed to formal analysis, investigation, and writing—review and editing. Qiuyun Xu, Ricardo Engel, Maria M. Tsoukas, and Steven Daveluy contributed to investigation, methodology, validation, and writing—review and editing. Darius Mehregan contributed to writing—review and editing. Kamran Avanaki contributed to formal analysis, methodology, resources, supervision, validation, and writing—original draft, review, and editing.

Acknowledgments

The authors would like to thank the Department of Dermatology at Wayne State University and the Dermatology Department at the University of Illinois at Chicago for their support. The authors also would like to thank the Melanoma Research Alliance (Grant No. 624320) for their support.

References

1. 

E. Linos et al., “Increasing burden of melanoma in the United States,” J. Invest. Dermatol., 129 (7), 1666 –1674 https://doi.org/10.1038/jid.2008.423 JIDEAE 0022-202X (2009). Google Scholar

2. 

C. L. Kosary et al., “Clinical and prognostic factors for melanoma of the skin using SEER registries: collaborative stage data collection system, version 1 and version 2,” Cancer, 120 3807 –3814 https://doi.org/10.1002/cncr.29050 CANCAR 0008-543X (2014). Google Scholar

3. 

Jr. G. P. Guy, “Vital signs: melanoma incidence and mortality trends and projections—United States, 1982–2030,” MMWR Morb. Mortal Wkly. Rep, 64 (21), 591 –596 (2015). Google Scholar

4. 

D. C. Whiteman, A. C. Green and C. M. Olsen, “The growing burden of invasive melanoma: projections of incidence rates and numbers of new cases in six susceptible populations through 2031,” J. Invest. Dermatol., 136 (6), 1161 –1171 https://doi.org/10.1016/j.jid.2016.01.035 JIDEAE 0022-202X (2016). Google Scholar

5. 

F. Erdmann et al., “International trends in the incidence of malignant melanoma 1953–2008—are recent generations at higher or lower risk?,” Int. J. Cancer, 132 (2), 385 –400 https://doi.org/10.1002/ijc.27616 IJCNAW 1097-0215 (2013). Google Scholar

6. 

A. Noone et al., “SEER cancer statistics review, 1975–2015,” (2018). Google Scholar

7. 

R. Shellenberger, M. Nabhan and S. Kakaraparthi, “Melanoma screening: a plan for improving early detection,” Ann. Med., 48 (3), 142 –148 https://doi.org/10.3109/07853890.2016.1145795 (2016). Google Scholar

9. 

L. Garibyan and D. E. Fisher, “How sunlight causes melanoma,” Curr. Oncol. Rep., 12 (5), 319 –326 https://doi.org/10.1007/s11912-010-0119-y (2010). Google Scholar

10. 

R. S. Stern, “Prevalence of a history of skin cancer in 2007: results of an incidence-based model,” Arch. Dermatol., 146 (3), 279 –282 https://doi.org/10.1001/archdermatol.2010.4 (2010). Google Scholar

11. 

S. M. Swetter et al., “Guidelines of care for the management of primary cutaneous melanoma,” J. Am. Acad. Dermatol., 80 (1), 208 –250 https://doi.org/10.1016/j.jaad.2018.08.055 JAADDB 0190-9622 (2019). Google Scholar

12. 

E. K. Bartlett and G. C. Karakousis, “Current staging and prognostic factors in melanoma,” Surg. Oncol. Clin. N. Am., 24 (2), 215 –227 https://doi.org/10.1016/j.soc.2014.12.001 (2015). Google Scholar

13. 

M. Lechner, C. Boshoff, S. Beck, “Cancer epigenome,” Advances in Genetics, 247 –276 Elsevier( (2010). Google Scholar

14. 

W. Weyers, “Screening for malignant melanoma-a critical assessment in historical perspective,” Dermatol. Pract. Conceptual, 8 (2), 89 –103 https://doi.org/10.5826/dpc.0802a06 (2018). Google Scholar

15. 

Y. Cheng et al., “Stage-specific prognostic biomarkers in melanoma,” Oncotarget, 6 (6), 4180 –4189 https://doi.org/10.18632/oncotarget.2907 (2015). Google Scholar

16. 

C. Fink and H. A. Haenssle, “Non-invasive tools for the diagnosis of cutaneous melanoma,” Skin Res. Technol., 23 (3), 261 –271 https://doi.org/10.1111/srt.12350 (2017). Google Scholar

17. 

M. A. Boone et al., “In vivo assessment of optical properties of melanocytic skin lesions and differentiation of melanoma from non-malignant lesions by high-definition optical coherence tomography,” Arch. Dermatol. Res., 308 (1), 7 –20 https://doi.org/10.1007/s00403-015-1608-5 (2016). Google Scholar

18. 

Q. Xu, The Advanced Applications for Optical Coherence Tomography in Skin Imaging, Wayne State University( (2021). Google Scholar

19. 

Z. Fayyaz et al., “A comparative study of optimization algorithms for wavefront shaping,” J. Innov. Opt. Health Sci., 12 (04), 1942002 https://doi.org/10.1142/S1793545819420021 (2019). Google Scholar

20. 

M. Forouzandeh et al., “The efficacy and morphological effects of hydrogen peroxide 40% topical solution for the treatment of seborrheic keratoses, evaluated by dynamic optical coherence tomography,” Skin Res. Technol., 26 (1), 142 –145 https://doi.org/10.1111/srt.12752 (2020). Google Scholar

21. 

E. Jalilian et al., “Contrast‐enhanced optical coherence tomography for melanoma detection: an in vitro study,” J. Biophotonics, 13 (5), e201960097 https://doi.org/10.1002/jbio.201960097 (2020). Google Scholar

22. 

M. R. Avanaki and A. Podoleanu, “En-face time-domain optical coherence tomography with dynamic focus for high-resolution imaging,” J. Biomed. Opt., 22 (5), 056009 https://doi.org/10.1117/1.JBO.22.5.056009 JBOPFO 1083-3668 (2017). Google Scholar

23. 

D. Tes et al., “Granular cell tumor imaging using optical coherence tomography,” Biomed. Eng. Comput. Biol., 9 1179597218790250 https://doi.org/10.1177/1179597218790250 (2018). Google Scholar

24. 

M. R. Avanaki et al., “Investigation of basal cell carcinoma using dynamic focus optical coherence tomography,” Appl. Opt., 52 (10), 2116 –2124 https://doi.org/10.1364/AO.52.002116 APOPAI 0003-6935 (2013). Google Scholar

25. 

M. R. Avanaki, A. Hojjat and A. G. Podoleanu, “Investigation of computer-based skin cancer detection using optical coherence tomography,” J. Mod. Opt., 56 (13), 1536 –1544 https://doi.org/10.1080/09500340902990007 JMOPEW 0950-0340 (2009). Google Scholar

26. 

Q. Xu et al., “Monitoring the topical delivery of ultrasmall gold nanoparticles using optical coherence tomography,” Skin Res. Technol., 26 (2), 263 –268 https://doi.org/10.1111/srt.12789 (2020). Google Scholar

27. 

J. Lee et al., “Noninvasive imaging exploration of phacomatosis pigmentokeratotica using high-frequency ultrasound and optical coherence tomography: can biopsy of PPK patients be avoided?,” Skin Res. Technol., 29 (4), e13279 https://doi.org/10.1111/srt.13279 (2023). Google Scholar

28. 

K. Avanaki and P. Andersen, “Oct radiomic features for differentiation of early malignant melanoma from benign nevus,” (2020). Google Scholar

29. 

J. Lee et al., “Optical coherence tomography confirms non‐malignant pigmented lesions in phacomatosis pigmentokeratotica using a support vector machine learning algorithm,” Skin Res. Technol., 29 (6), e13377 https://doi.org/10.1111/srt.13377 (2023). Google Scholar

30. 

K. Avanaki, P. E. Andersen, “Optical coherence tomography for melanoma detection,” New Technologies in Dermatological Science and Practice, 47 –58 CRC Press( (2021). Google Scholar

31. 

A. Rajabi-Estarabadi et al., “Optical coherence tomography imaging of melanoma skin cancer,” Lasers Med. Sci., 34 411 –420 https://doi.org/10.1007/s10103-018-2696-1 LMSCEZ 1435-604X (2019). Google Scholar

32. 

Z. Turani et al., “Optical radiomic signatures derived from OCT images to improve identification of melanoma,” in Eur. Conf. Biomed. Opt., (2019). Google Scholar

33. 

B. Lukas et al., “Skin cancer detection using optical coherence tomography,” Optical Spectroscopy and Imaging for Cancer Diagnostics: Fundamentals, Progress, and Challenges, 473 –495 World Scientific( (2023). Google Scholar

34. 

Q. Xu et al., “Swept-source optical coherence tomography–supervised biopsy,” Dermatol. Surg., 44 (6), 768 –775 https://doi.org/10.1097/DSS.0000000000001475 (2018). Google Scholar

35. 

S. Adabi et al., “Universal in vivo textural model for human skin based on optical coherence tomograms,” Sci. Rep., 7 (1), 17912 https://doi.org/10.1038/s41598-017-17398-8 (2017). Google Scholar

36. 

C. Herman, “Emerging technologies for the detection of melanoma: achieving better outcomes,” Clin. Cosmet. Investig. Dermatol., 5 195 https://doi.org/10.2147/CCID.S27902 (2012). Google Scholar

37. 

R. Manwar et al., “Deep learning protocol for improved photoacoustic brain imaging,” J. Biophotonics, 13 (10), e202000212 https://doi.org/10.1002/jbio.202000212 (2020). Google Scholar

38. 

K. Kratkiewicz et al., “Technical considerations in the Verasonics research ultrasound platform for developing a photoacoustic imaging system,” Biomed. Opt. Express, 12 (2), 1050 –1084 https://doi.org/10.1364/BOE.415481 BOEICL 2156-7085 (2021). Google Scholar

39. 

K. Kratkiewicz et al., “Photoacoustic/ultrasound/optical coherence tomography evaluation of melanoma lesion and healthy skin in a swine model,” Sensors, 19 (12), 2815 https://doi.org/10.3390/s19122815 SNSRES 0746-9462 (2019). Google Scholar

40. 

A. Hariri et al., “Towards low cost photoacoustic microscopy system for evaluation of skin health,” Proc. SPIE, 9976 99760X https://doi.org/10.1117/12.2238423 PSISDG 0277-786X (2016). Google Scholar

41. 

A. Blundo et al., “Comparative analysis of diagnostic techniques for melanoma detection: a systematic review of diagnostic test accuracy studies and meta-analysis,” Front. Med., 8 637069 https://doi.org/10.3389/fmed.2021.637069 (2021). Google Scholar

42. 

Y. Xing et al., “Contemporary diagnostic imaging modalities for the staging and surveillance of melanoma patients: a meta-analysis,” J. Natl. Cancer Inst., 103 (2), 129 –142 https://doi.org/10.1093/jnci/djq455 JNCIEQ (2011). Google Scholar

43. 

Y. Xing, K. D. Cromwell and J. N. Cormier, “Review of diagnostic imaging modalities for the surveillance of melanoma patients,” Dermatol. Res. Pract., 2012 941921 https://doi.org/10.1155/2012/941921 (2012). Google Scholar

44. 

I. Steinberg et al., “Photoacoustic clinical imaging,” Photoacoustics, 14 77 –98 https://doi.org/10.1016/j.pacs.2019.05.001 (2019). Google Scholar

45. 

M. Liu and W. Drexler, “Optical coherence tomography angiography and photoacoustic imaging in dermatology,” Photochem. Photobiol. Sci., 18 (5), 945 –962 https://doi.org/10.1039/c8pp00471d (2019). Google Scholar

46. 

T. Vienneau, T. Vu, J. Yao, “Photoacoustic imaging of skin,” Imaging Technologies and Transdermal Delivery in Skin Disorders, 411 –442 (2019). Google Scholar

47. 

P. Beard, “Biomedical photoacoustic imaging,” Interface Focus, 1 (4), 602 –631 https://doi.org/10.1098/rsfs.2011.0028 (2011). Google Scholar

48. 

M. W. Schellenberg and H. K. Hunt, “Hand-held optoacoustic imaging: a review,” Photoacoustics, 11 14 –27 https://doi.org/10.1016/j.pacs.2018.07.001 (2018). Google Scholar

49. 

S. Liu et al., “Continuous wave laser excitation based portable optoacoustic imaging system for melanoma detection,” in IEEE Biomed. Circuits and Syst. Conf. (BioCAS), (2019). https://doi.org/10.1109/BIOCAS.2019.8919157 Google Scholar

50. 

M. Nasiriavanaki et al., “High-resolution photoacoustic tomography of resting-state functional connectivity in the mouse brain,” Proc. Natl. Acad. Sci. U. S. A., 111 (1), 21 –26 https://doi.org/10.1073/pnas.1311868111 (2014). Google Scholar

51. 

N. Meimani et al., “A numerical analysis of a semi-dry coupling configuration in photoacoustic computed tomography for infant brain imaging,” Photoacoustics, 7 27 –35 https://doi.org/10.1016/j.pacs.2017.06.001 (2017). Google Scholar

52. 

D. Nasri et al., “Photoacoustic imaging for investigating tumor hypoxia: a strategic assessment,” Theranostics, 13 (10), 3346 https://doi.org/10.7150/thno.84253 (2023). Google Scholar

53. 

A. Hariri et al., “Development of low-cost photoacoustic imaging systems using very low-energy pulsed laser diodes,” J. Biomed. Opt., 22 (7), 075001 https://doi.org/10.1117/1.JBO.22.7.075001 JBOPFO 1083-3668 (2017). Google Scholar

54. 

S. Mahmoodkalayeh et al., “Low temperature-mediated enhancement of photoacoustic imaging depth,” Sci. Rep., 8 (1), 4873 https://doi.org/10.1038/s41598-018-22898-2 (2018). Google Scholar

55. 

L. Mohammadi et al., “Skull’s acoustic attenuation and dispersion modeling on photoacoustic signal,” Proc. SPIE, 10494 104946K https://doi.org/10.1117/12.2291362 PSISDG 0277-786X (2018). Google Scholar

56. 

L. Mohammadi et al., “Skull’s aberration modeling: towards photoacoustic human brain imaging,” Proc. SPIE, 10878 108785W https://doi.org/10.1117/12.2510549 PSISDG 0277-786X (2019). Google Scholar

57. 

A.-R. Mohammadi-Nejad et al., “Neonatal brain resting-state functional connectivity imaging modalities,” Photoacoustics, 10 1 –19 https://doi.org/10.1016/j.pacs.2018.01.003 (2018). Google Scholar

58. 

M. Mozaffarzadeh et al., “Double-stage delay multiply and sum beamforming algorithm: application to linear-array photoacoustic imaging,” IEEE Trans. Biomed. Eng., 65 (1), 31 –42 https://doi.org/10.1109/TBME.2017.2690959 IEBEAX 0018-9294 (2018). Google Scholar

59. 

Y. Zhou et al., “A phosphorus phthalocyanine formulation with intense absorbance at 1000 nm for deep optical imaging,” Theranostics, 6 (5), 688 https://doi.org/10.7150/thno.14555 (2016). Google Scholar

60. 

C. Benvenuto-Andrade et al., “Differences between polarized light dermoscopy and immersion contact dermoscopy for the evaluation of skin lesions,” Arch. Dermatol., 143 (3), 329 –338 https://doi.org/10.1001/archderm.143.3.329 (2007). Google Scholar

61. 

S. Segura et al., “Development of a two-step method for the diagnosis of melanoma by reflectance confocal microscopy,” J. Am. Acad. Dermatol., 61 (2), 216 –229 https://doi.org/10.1016/j.jaad.2009.02.014 JAADDB 0190-9622 (2009). Google Scholar

62. 

T. Gambichler et al., “Characterization of benign and malignant melanocytic skin lesions using optical coherence tomography in vivo,” J. Am. Acad. Dermatol., 57 (4), 629 –637 https://doi.org/10.1016/j.jaad.2007.05.029 JAADDB 0190-9622 (2007). Google Scholar

63. 

M. H. Eybposh et al., “Cluster-based filtering framework for speckle reduction in OCT images,” Biomed. Opt. Express, 9 (12), 6359 –6373 https://doi.org/10.1364/BOE.9.006359 BOEICL 2156-7085 (2018). Google Scholar

64. 

S. Hojjatoleslami, M. Avanaki and A. G. Podoleanu, “Image quality improvement in optical coherence tomography using Lucy–Richardson deconvolution algorithm,” Appl. Opt., 52 (23), 5663 –5670 https://doi.org/10.1364/AO.52.005663 APOPAI 0003-6935 (2013). Google Scholar

65. 

S. Adabi et al., “Learnable despeckling framework for optical coherence tomography images,” J. Biomed. Opt., 23 (1), 016013 https://doi.org/10.1117/1.JBO.23.1.016013 JBOPFO 1083-3668 (2018). Google Scholar

66. 

Z. Turani et al., “Noise reduction in OCT skin images,” Proc. SPIE, 10137 1013724 https://doi.org/10.1117/12.2255548 PSISDG 0277-786X (2017). Google Scholar

67. 

S. O’leary et al., “OCT image atlas of healthy skin on sun-exposed areas,” Skin Res. Technol., 24 (4), 570 –586 https://doi.org/10.1111/srt.12468 (2018). Google Scholar

68. 

A. Hojjatoleslami and M. R. Avanaki, “OCT skin image enhancement through attenuation compensation,” Appl. Opt., 51 (21), 4927 –4935 https://doi.org/10.1364/AO.51.004927 APOPAI 0003-6935 (2012). Google Scholar

69. 

M.-R. Nasiri-Avanaki et al., “Optical coherence tomography system optimization using simulated annealing algorithm,” in Proc. Math. Methods and Appl. Comput., (2009). Google Scholar

70. 

S. Adabi et al., “Optical coherence tomography technology and quality improvement methods for optical coherence tomography images of skin: a short review,” Biomed. Eng. Comput. Biol., 8 117959721771347 https://doi.org/10.1177/1179597217713475 (2017). Google Scholar

71. 

S. Adabi et al., “An overview of methods to mitigate artifacts in optical coherence tomography imaging of the skin,” Skin Res. Technol., 24 (2), 265 –273 https://doi.org/10.1111/srt.12423 (2018). Google Scholar

72. 

Z. Turani et al., “Refractive index correction in optical coherence tomography images of multilayer tissues,” J. Biomed. Opt., 23 (7), 070501 https://doi.org/10.1117/1.JBO.23.7.070501 JBOPFO 1083-3668 (2018). Google Scholar

73. 

M. R. Avanaki and A. Hojjatoleslami, “Skin layer detection of optical coherence tomography images,” Optik, 124 (22), 5665 –5668 https://doi.org/10.1016/j.ijleo.2013.04.033 OTIKAJ 0030-4026 (2013). Google Scholar

74. 

M. R. Avanaki et al., “Spatial compounding algorithm for speckle reduction of dynamic focus OCT images,” IEEE Photonics Technol. Lett., 25 (15), 1439 –1442 https://doi.org/10.1109/LPT.2013.2266660 IPTLEL 1041-1135 (2013). Google Scholar

75. 

M. R. Avanaki et al., “Speckle reduction using an artificial neural network algorithm,” Appl. Opt., 52 (21), 5050 –5057 https://doi.org/10.1364/AO.52.005050 APOPAI 0003-6935 (2013). Google Scholar

76. 

J. May et al., “40638 Comprehensive Atlas of healthy skin using optical coherence tomography,” J. Am. Acad. Dermatol., 89 (3), AB140 https://doi.org/10.1016/j.jaad.2023.07.563 JAADDB 0190-9622 (2023). Google Scholar

77. 

Q. Xu et al., “Study of variation of attenuation coefficient in different skin color and age groups using optical coherence tomography,” Proc. SPIE, PC11934 PC119340P https://doi.org/10.1117/12.2613007 PSISDG 0277-786X (2022). Google Scholar

78. 

A. Taghavikhalilbad et al., “Semi-automated localization of dermal epidermal junction in optical coherence tomography images of skin,” Appl. Opt., 56 (11), 3116 –3121 https://doi.org/10.1364/AO.56.003116 APOPAI 0003-6935 (2017). Google Scholar

79. 

M. R. Avanaki et al., “Two applications of solid phantoms in performance assessment of optical coherence tomography systems,” Appl. Opt., 52 (29), 7054 –7061 https://doi.org/10.1364/AO.52.007054 APOPAI 0003-6935 (2013). Google Scholar

80. 

M. Crisan et al., “Ultrasonographic staging of cutaneous malignant tumors: an ultrasonographic depth index,” Arch. Dermatol. Res., 305 (4), 305 –313 https://doi.org/10.1007/s00403-013-1321-1 (2013). Google Scholar

81. 

M.-H. Schmid-Wendtner and D. Dill-Müller, “Ultrasound technology in dermatology,” Semin. Cutan. Med. Surg., 27 (1), 44 –51 https://doi.org/10.1016/j.sder.2008.01.003 (2008). Google Scholar

82. 

M. Omar, J. Aguirre and V. Ntziachristos, “Optoacoustic mesoscopy for biomedicine,” Nat. Biomed. Eng., 3 (5), 354 –370 https://doi.org/10.1038/s41551-019-0377-4 (2019). Google Scholar

83. 

L. V. Wang, “Multiscale photoacoustic microscopy and computed tomography,” Nat. Photonics, 3 (9), 503 –509 https://doi.org/10.1038/nphoton.2009.157 NPAHBY 1749-4885 (2009). Google Scholar

84. 

M. Schwarz et al., “Implications of ultrasound frequency in optoacoustic mesoscopy of the skin,” IEEE Trans. Med. Imaging, 34 (2), 672 –677 https://doi.org/10.1109/TMI.2014.2365239 ITMID4 0278-0062 (2014). Google Scholar

85. 

D. Piras et al., “Photoacoustic imaging of the breast using the twente photoacoustic mammoscope: present status and future perspectives,” IEEE J. Sel. Top. Quantum Electron., 16 (4), 730 –739 https://doi.org/10.1109/JSTQE.2009.2034870 IJSQEN 1077-260X (2009). Google Scholar

86. 

F. Knieling et al., “Multispectral optoacoustic tomography for assessment of Crohn’s disease activity,” N. Engl. J. Med., 376 (13), 1292 –1294 https://doi.org/10.1056/NEJMc1612455 NEJMBH (2017). Google Scholar

87. 

J. Reber et al., “Non-invasive measurement of brown fat metabolism based on optoacoustic imaging of hemoglobin gradients,” Cell Metab., 27 (3), 689 –701.e4 https://doi.org/10.1016/j.cmet.2018.02.002 1550-4131 (2018). Google Scholar

88. 

A. Dima and V. Ntziachristos, “Non-invasive carotid imaging using optoacoustic tomography,” Opt. Express, 20 (22), 25044 –25057 https://doi.org/10.1364/OE.20.025044 OPEXFF 1094-4087 (2012). Google Scholar

89. 

M. P. Fronheiser et al., “Real-time optoacoustic monitoring and three-dimensional mapping of a human arm vasculature,” J. Biomed. Opt., 15 (2), 021305 https://doi.org/10.1117/1.3370336 JBOPFO 1083-3668 (2010). Google Scholar

90. 

A. Buehler et al., “Real-time handheld multispectral optoacoustic imaging,” Opt. Lett., 38 (9), 1404 –1406 https://doi.org/10.1364/OL.38.001404 OPLEDP 0146-9592 (2013). Google Scholar

91. 

M. Xu and L. V. Wang, “Photoacoustic imaging in biomedicine,” Rev. Sci. Instrum., 77 (4), 041101 https://doi.org/10.1063/1.2195024 RSINAK 0034-6748 (2006). Google Scholar

92. 

A. Taruttis and V. Ntziachristos, “Advances in real-time multispectral optoacoustic imaging and its applications,” Nat. Photonics, 9 (4), 219 –227 https://doi.org/10.1038/nphoton.2015.29 NPAHBY 1749-4885 (2015). Google Scholar

93. 

V. Ntziachristos, “Going deeper than microscopy: the optical imaging frontier in biology,” Nat. Methods, 7 (8), 603 –614 https://doi.org/10.1038/nmeth.1483 1548-7091 (2010). Google Scholar

94. 

E. M. Strohm, E. S. Berndl and M. C. Kolios, “High frequency label-free photoacoustic microscopy of single cells,” Photoacoustics, 1 (3–4), 49 –53 https://doi.org/10.1016/j.pacs.2013.08.003 (2013). Google Scholar

95. 

M.-L. Li et al., “Improved in vivo photoacoustic microscopy based on a virtual-detector concept,” Opt. Lett., 31 (4), 474 –476 https://doi.org/10.1364/OL.31.000474 OPLEDP 0146-9592 (2006). Google Scholar

96. 

A. Danielli et al., “Label-free photoacoustic nanoscopy,” J. Biomed. Opt., 19 (8), 086006 https://doi.org/10.1117/1.JBO.19.8.086006 JBOPFO 1083-3668 (2014). Google Scholar

97. 

B. Hindelang et al., “Non-invasive imaging in dermatology and the unique potential of raster-scan optoacoustic mesoscopy,” J. Eur. Acad. Dermatol. Venereol., 33 (6), 1051 –1061 https://doi.org/10.1111/jdv.15342 JEAVEQ 0926-9959 (2019). Google Scholar

98. 

V. Ntziachristos et al., “Emerging technologies to image tissue metabolism,” Cell Metab., 29 (3), 518 –538 https://doi.org/10.1016/j.cmet.2018.09.004 1550-4131 (2019). Google Scholar

99. 

V. Ntziachristos and D. Razansky, “Molecular imaging by means of multispectral optoacoustic tomography (MSOT),” Chem. Rev., 110 (5), 2783 –2794 https://doi.org/10.1021/cr9002566 CHREAY 0009-2665 (2010). Google Scholar

100. 

Y. W. Yew et al., “Investigation of morphological, vascular and biochemical changes in the skin of an atopic dermatitis (AD) patient in response to dupilumab using raster scanning optoacoustic mesoscopy (RSOM) and handheld confocal Raman spectroscopy (CRS),” J. Dermatol. Sci., 95 (3), 123 –125 https://doi.org/10.1016/j.jdermsci.2019.07.003 JDSCEI 0923-1811 (2019). Google Scholar

101. 

J. Aguirre et al., “Assessing nailfold microvascular structure with ultra-wideband raster-scan optoacoustic mesoscopy,” Photoacoustics, 10 31 –37 https://doi.org/10.1016/j.pacs.2018.02.002 (2018). Google Scholar

102. 

A. Berezhnoi et al., “Assessing hyperthermia-induced vasodilation in human skin in vivo using optoacoustic mesoscopy,” J. Biophotonics, 11 (11), e201700359 https://doi.org/10.1002/jbio.201700359 (2018). Google Scholar

103. 

J. Hult et al., “Comparison of photoacoustic imaging and histopathological examination in determining the dimensions of 52 human melanomas and nevi ex vivo,” Biomed. Opt. Express, 12 (7), 4097 –4114 https://doi.org/10.1364/BOE.425524 BOEICL 2156-7085 (2021). Google Scholar

104. 

C. Lee et al., “Three-dimensional clinical handheld photoacoustic/ultrasound scanner,” Photoacoustics, 18 100173 https://doi.org/10.1016/j.pacs.2020.100173 (2020). Google Scholar

105. 

V. Neuschmelting et al., “Performance of a multispectral optoacoustic tomography (MSOT) system equipped with 2D vs. 3D handheld probes for potential clinical translation,” Photoacoustics, 4 (1), 1 –10 https://doi.org/10.1016/j.pacs.2015.12.001 (2016). Google Scholar

106. 

C. Kim et al., “Deeply penetrating in vivo photoacoustic imaging using a clinical ultrasound array system,” Biomed. Opt. Express, 1 (1), 278 –284 https://doi.org/10.1364/BOE.1.000278 BOEICL 2156-7085 (2010). Google Scholar

107. 

J. Aguirre et al., “Broadband mesoscopic optoacoustic tomography reveals skin layers,” Opt. Lett., 39 (21), 6297 –6300 https://doi.org/10.1364/OL.39.006297 OPLEDP 0146-9592 (2014). Google Scholar

108. 

L. V. Wang and S. Hu, “Photoacoustic tomography: in vivo imaging from organelles to organs,” Science, 335 (6075), 1458 –1462 https://doi.org/10.1126/science.1216210 SCIEAS 0036-8075 (2012). Google Scholar

109. 

H. He et al., “Fast raster-scan optoacoustic mesoscopy enables assessment of human melanoma microvasculature in vivo,” Nat. Commun., 13 (1), 2803 https://doi.org/10.1038/s41467-022-30471-9 NCAOBW 2041-1723 (2022). Google Scholar

110. 

J. Yao and L. Wang, “Photoacoustic microscopy,” Laser Photonics Rev., 7 (5), 758 –778 https://doi.org/10.1002/lpor.201200060 (2013). Google Scholar

111. 

J. Yao et al., “Label-free oxygen-metabolic photoacoustic microscopy in vivo,” J. Biomed. Opt., 16 (7), 076003 https://doi.org/10.1117/1.3594786 JBOPFO 1083-3668 (2011). Google Scholar

112. 

C. Zhang et al., “Multiscale high-speed photoacoustic microscopy based on free-space light transmission and a MEMS scanning mirror,” Opt. Lett., 45 (15), 4312 –4315 https://doi.org/10.1364/OL.397733 OPLEDP 0146-9592 (2020). Google Scholar

113. 

H. F. Zhang et al., “Functional photoacoustic microscopy for high-resolution and noninvasive in vivo imaging,” Nat. Biotechnol., 24 (7), 848 –851 https://doi.org/10.1038/nbt1220 NABIF9 1087-0156 (2006). Google Scholar

114. 

J. W. Baik et al., “Super wide-field photoacoustic microscopy of animals and humans in vivo,” IEEE Trans. Med. Imaging, 39 (4), 975 –984 https://doi.org/10.1109/TMI.2019.2938518 ITMID4 0278-0062 (2019). Google Scholar

115. 

A. Breathnach et al., “Preoperative measurement of cutaneous melanoma and nevi thickness with photoacoustic imaging,” J. Med. Imaging, 5 (1), 015004 https://doi.org/10.1117/1.JMI.5.1.015004 JMEIET 0920-5497 (2018). Google Scholar

116. 

J. T. Oh et al., “Three-dimensional imaging of skin melanoma in vivo by dual-wavelength photoacoustic microscopy,” J. Biomed. Opt., 11 (3), 034032 https://doi.org/10.1117/1.2210907 JBOPFO 1083-3668 (2006). Google Scholar

117. 

I. Stoffels et al., “Metastatic status of sentinel lymph nodes in melanoma determined noninvasively with multispectral optoacoustic imaging,” Sci. Transl. Med., 7 (317), 317ra199 https://doi.org/10.1126/scitranslmed.aad1278 STMCBQ 1946-6234 (2015). Google Scholar

118. 

A. Dima, N. C. Burton and V. Ntziachristos, “Multispectral optoacoustic tomography at 64, 128, and 256 channels,” J. Biomed. Opt., 19 (3), 036021 https://doi.org/10.1117/1.JBO.19.3.036021 JBOPFO 1083-3668 (2014). Google Scholar

119. 

S. Chuah et al., “Structural and functional 3D mapping of skin tumours with non‐invasive multispectral optoacoustic tomography,” Skin Res. Technol., 23 (2), 221 –226 https://doi.org/10.1111/srt.12326 (2017). Google Scholar

120. 

M. Mehrmohammadi et al., “Photoacoustic imaging for cancer detection and staging,” Curr. Mol. Imaging (Discont.), 2 (1), 89 –105 https://doi.org/10.2174/2211555211302010010 (2013). Google Scholar

121. 

M. R. Tomaszewski et al., “Oxygen enhanced optoacoustic tomography (OE-OT) reveals vascular dynamics in murine models of prostate cancer,” Theranostics, 7 (11), 2900 https://doi.org/10.7150/thno.19841 (2017). Google Scholar

122. 

G. Balasundaram et al., “Noninvasive anatomical and functional imaging of orthotopic glioblastoma development and therapy using multispectral optoacoustic tomography,” Transl. Oncol., 11 (5), 1251 –1258 https://doi.org/10.1016/j.tranon.2018.07.001 (2018). Google Scholar

123. 

N. C. Burton et al., “Multispectral opto-acoustic tomography (MSOT) of the brain and glioblastoma characterization,” Neuroimage, 65 522 –528 https://doi.org/10.1016/j.neuroimage.2012.09.053 NEIMEF 1053-8119 (2013). Google Scholar

124. 

A. Samykutty et al., “Optoacoustic imaging identifies ovarian cancer using a microenvironment targeted theranostic wormhole mesoporous silica nanoparticle,” Biomaterials, 182 114 –126 https://doi.org/10.1016/j.biomaterials.2018.08.001 BIMADU 0142-9612 (2018). Google Scholar

125. 

A. Dima and V. Ntziachristos, “In-vivo handheld optoacoustic tomography of the human thyroid,” Photoacoustics, 4 (2), 65 –69 https://doi.org/10.1016/j.pacs.2016.05.003 (2016). Google Scholar

126. 

W. Roll et al., “Multispectral optoacoustic tomography of benign and malignant thyroid disorders: a pilot study,” J. Nucl. Med., 60 (10), 1461 –1466 https://doi.org/10.2967/jnumed.118.222174 JNMEAQ 0161-5505 (2019). Google Scholar

127. 

Y. Wu et al., “Activatable probes for diagnosing and positioning liver injury and metastatic tumors by multispectral optoacoustic tomography,” Nat. Commun., 9 (1), 3983 https://doi.org/10.1038/s41467-018-06499-1 NCAOBW 2041-1723 (2018). Google Scholar

128. 

I. Ivankovic et al., “Volumetric optoacoustic tomography enables non-invasive in vivo characterization of impaired heart function in hypoxic conditions,” Sci. Rep., 9 (1), 1 –8 https://doi.org/10.1038/s41598-019-44818-8 (2019). Google Scholar

129. 

M. Masthoff et al., “Multispectral optoacoustic tomography of systemic sclerosis,” J. Biophotonics, 11 (11), e201800155 https://doi.org/10.1002/jbio.201800155 (2018). Google Scholar

130. 

A. B. E. Attia et al., “Noninvasive real-time characterization of non-melanoma skin cancers with handheld optoacoustic probes,” Photoacoustics, 7 20 –26 https://doi.org/10.1016/j.pacs.2017.05.003 (2017). Google Scholar

131. 

S. Y. Chuah et al., “Volumetric multispectral optoacoustic tomography for 3-dimensional reconstruction of skin tumors: a further evaluation with histopathologic correlation,” J. Invest. Dermatol., 139 (2), 481 –485 https://doi.org/10.1016/j.jid.2018.08.014 (2019). Google Scholar

132. 

W. Zhou et al., “Optical biopsy approach to basal cell carcinoma and melanoma based on all-optically integrated photoacoustic and optical coherence tomography,” Opt. Lett., 42 (11), 2145 –2148 https://doi.org/10.1364/OL.42.002145 OPLEDP 0146-9592 (2017). Google Scholar

133. 

B. Park et al., “3D wide-field multispectral photoacoustic imaging of human melanomas in vivo: a pilot study,” J. Eur. Acad. Dermatol. Venereol., 35 (3), 669 –676 https://doi.org/10.1111/jdv.16985 JEAVEQ 0926-9959 (2021). Google Scholar

134. 

Z. Xu et al., “Visualizing tumor angiogenesis and boundary with polygon-scanning multiscale photoacoustic microscopy,” Photoacoustics, 26 100342 https://doi.org/10.1016/j.pacs.2022.100342 (2022). Google Scholar

135. 

Y. Zhou et al., “Noninvasive determination of melanoma depth using a handheld photoacoustic probe,” J. Invest. Dermatol., 137 (6), 1370 https://doi.org/10.1016/j.jid.2017.01.016 (2017). Google Scholar

136. 

S.-W. Cho et al., “Efficient label-free in vivo photoacoustic imaging of melanoma cells using a condensed NIR-I spectral window,” Photoacoustics, 29 100456 https://doi.org/10.1016/j.pacs.2023.100456 (2023). Google Scholar

137. 

J. A. Viator et al., “Photoacoustic detection of circulating melanoma cells in late stage patients,” J. Innov. Opt. Health Sci., 13 (06), 2050023 https://doi.org/10.1142/S1793545820500236 (2020). Google Scholar

138. 

O. V. Grishin et al., “Effect of pulsed laser parameters on photoacoustic flow cytometry efficiency in vitro and in vivo,” Cytometry Part A, 103 868 –880 https://doi.org/10.1002/cyto.a.24778 1552-4922 (2023). Google Scholar

139. 

L. Li et al., “Single-impulse panoramic photoacoustic computed tomography of small-animal whole-body dynamics at high spatiotemporal resolution,” Nat. Biomed. Eng., 1 (5), 0071 https://doi.org/10.1038/s41551-017-0071 (2017). Google Scholar

140. 

Y. Wang et al., “Fiber-laser-based photoacoustic microscopy and melanoma cell detection,” J. Biomed. Opt., 16 (1), 011014 https://doi.org/10.1117/1.3525643 JBOPFO 1083-3668 (2011). Google Scholar

141. 

M. Han et al., “In vivo dual-modal photoacoustic and ultrasound imaging of sentinel lymph nodes using a solid-state dye laser system,” Sensors, 20 (13), 3714 https://doi.org/10.3390/s20133714 SNSRES 0746-9462 (2020). Google Scholar

142. 

M. T. Martell et al., “Deep learning-enabled realistic virtual histology with ultraviolet photoacoustic remote sensing microscopy,” Nat. Commun., 14 (1), 5967 https://doi.org/10.1038/s41467-023-41574-2 NCAOBW 2041-1723 (2023). Google Scholar

143. 

L. Nie and X. Chen, “Structural and functional photoacoustic molecular tomography aided by emerging contrast agents,” Chem. Soc. Rev., 43 (20), 7132 –7170 https://doi.org/10.1039/C4CS00086B CSRVBR 0306-0012 (2014). Google Scholar

144. 

P. K. Upputuri and M. Pramanik, “Recent advances in photoacoustic contrast agents for in vivo imaging,” WIREs Nanomed. Nanobiotechnol., 12 (4), e1618 https://doi.org/10.1002/wnan.1618 (2020). Google Scholar

145. 

A. B. E. Attia et al., “A review of clinical photoacoustic imaging: current and future trends,” Photoacoustics, 16 100144 https://doi.org/10.1016/j.pacs.2019.100144 (2019). Google Scholar

146. 

B. Cox et al., “Quantitative spectroscopic photoacoustic imaging: a review,” J. Biomed. Opt., 17 (6), 061202 https://doi.org/10.1117/1.JBO.17.6.061202 JBOPFO 1083-3668 (2012). Google Scholar

147. 

M. Erfanzadeh and Q. Zhu, “Photoacoustic imaging with low-cost sources; a review,” Photoacoustics, 14 1 –11 https://doi.org/10.1016/j.pacs.2019.01.004 (2019). Google Scholar

148. 

J. Gröhl et al., “Deep learning for biomedical photoacoustic imaging: a review,” Photoacoustics, 22 100241 https://doi.org/10.1016/j.pacs.2021.100241 (2021). Google Scholar

149. 

S. Jeon et al., “Review on practical photoacoustic microscopy,” Photoacoustics, 15 100141 https://doi.org/10.1016/j.pacs.2019.100141 (2019). Google Scholar

150. 

T. D. Le, S.-Y. Kwon and C. Lee, “Segmentation and quantitative analysis of photoacoustic imaging: a review,” Photonics, 9 176 https://doi.org/10.3390/photonics9030176 (2022). Google Scholar

151. 

H. Lee et al., “Review on ultrasound-guided photoacoustic imaging for complementary analyses of biological systems in vivo,” Exp. Biol. Med. (Maywood), 248 (9), 762 –774 https://doi.org/10.1177/15353702231181341 EXBMAA 0071-3384 (2023). Google Scholar

152. 

L. Lin and L. V. Wang, “The emerging role of photoacoustic imaging in clinical oncology,” Nat. Rev. Clin. Oncol., 19 (6), 365 –384 https://doi.org/10.1038/s41571-022-00615-3 (2022). Google Scholar

153. 

S. Manohar and S. Gambhir, “Clinical photoacoustic imaging,” Photoacoustics, 19 100196 https://doi.org/10.1016/j.pacs.2020.100196 (2020). Google Scholar

154. 

D. Ren et al., “A review of transparent sensors for photoacoustic imaging applications,” Photonics, 8 324 https://doi.org/10.3390/photonics8080324 (2021). Google Scholar

155. 

J. J. Riksen, A. V. Nikolaev and G. van Soest, “Photoacoustic imaging on its way toward clinical utility: a tutorial review focusing on practical application in medicine,” J. Biomed. Opt., 28 (12), 121205 https://doi.org/10.1117/1.JBO.28.12.121205 JBOPFO 1083-3668 (2023). Google Scholar

156. 

P. K. Upputuri and M. Pramanik, “Fast photoacoustic imaging systems using pulsed laser diodes: a review,” Biomed. Eng. Lett., 8 (2), 167 –181 https://doi.org/10.1007/s13534-018-0060-9 (2018). Google Scholar

157. 

P. K. Upputuri and M. Pramanik, “Photoacoustic imaging in the second near-infrared window: a review,” J. Biomed. Opt., 24 (4), 040901 https://doi.org/10.1117/1.JBO.24.4.040901 JBOPFO 1083-3668 (2019). Google Scholar

158. 

T. von Knorring and M. Mogensen, “Photoacoustic tomography for assessment and quantification of cutaneous and metastatic malignant melanoma-A systematic review,” Photodiagn. Photodyn. Ther., 33 102095 https://doi.org/10.1016/j.pdpdt.2020.102095 (2021). Google Scholar

159. 

X. Yang et al., “Photoacoustic imaging for monitoring of stroke diseases: a review,” Photoacoustics, 23 100287 https://doi.org/10.1016/j.pacs.2021.100287 (2021). Google Scholar

160. 

J. Zhou and J. V. Jokerst, “Photoacoustic imaging with fiber optic technology: a review,” Photoacoustics, 20 100211 https://doi.org/10.1016/j.pacs.2020.100211 (2020). Google Scholar

161. 

Y. Zhu et al., “Towards clinical translation of LED-based photoacoustic imaging: a review,” Sensors, 20 (9), 2484 https://doi.org/10.3390/s20092484 SNSRES 0746-9462 (2020). Google Scholar

162. 

D. Li et al., “Seeing through the skin: photoacoustic tomography of skin vasculature and beyond,” JID Innov., 1 (3), 100039 https://doi.org/10.1016/j.xjidi.2021.100039 (2021). Google Scholar

163. 

J. Yao and L. V. Wang, “Photoacoustic tomography: fundamentals, advances and prospects,” Contrast Media Mol. Imaging, 6 (5), 332 –345 https://doi.org/10.1002/cmmi.443 (2011). Google Scholar

164. 

M. Schwarz et al., “Optoacoustic imaging of skin,” Imaging in Dermatology, 375 –385 Elsevier( (2016). Google Scholar

165. 

E. I. Galanzha et al., “In vivo fiber-based multicolor photoacoustic detection and photothermal purging of metastasis in sentinel lymph nodes targeted by nanoparticles,” J. Biophotonics, 2 (8–9), 528 –539 https://doi.org/10.1002/jbio.200910046 (2009). Google Scholar

166. 

E. I. Galanzha et al., “In vivo liquid biopsy using Cytophone platform for photoacoustic detection of circulating tumor cells in patients with melanoma,” Sci. Transl. Med., 11 (496), eaat5857 https://doi.org/10.1126/scitranslmed.aat5857 (2019). Google Scholar

167. 

E. I. Galanzha et al., “In vivo, noninvasive, label-free detection and eradication of circulating metastatic melanoma cells using two-color photoacoustic flow cytometry with a diode laser,” Cancer Res., 69 (20), 7926 –7934 https://doi.org/10.1158/0008-5472.CAN-08-4900 CNREA8 0008-5472 (2009). Google Scholar

168. 

E. I. Galanzha et al., “In vivo multispectral, multiparameter, photoacoustic lymph flow cytometry with natural cell focusing, label-free detection and multicolor nanoparticle probes,” Cytometry Part A: J. Int. Soc. Anal. Cytol., 73 (10), 884 –894 https://doi.org/10.1002/cyto.a.20587 (2008). Google Scholar

169. 

D. J. Grootendorst et al., “First experiences of photoacoustic imaging for detection of melanoma metastases in resected human lymph nodes,” Lasers Surg. Med., 44 (7), 541 –549 https://doi.org/10.1002/lsm.22058 LSMEDI 0196-8092 (2012). Google Scholar

170. 

G. Gutierrez-Juarez et al., “Optical photoacoustic detection of circulating melanoma cells in vitro,” Int. J. Thermophys., 31 (4), 784 –792 https://doi.org/10.1007/s10765-010-0770-4 IJTHDY 0195-928X (2010). Google Scholar

171. 

P. Hai et al., “Label-free high-throughput detection and quantification of circulating melanoma tumor cell clusters by linear-array-based photoacoustic tomography,” J. Biomed. Opt., 22 (4), 041004 https://doi.org/10.1117/1.JBO.22.4.041004 JBOPFO 1083-3668 (2016). Google Scholar

172. 

Y. He et al., “In vivo label-free photoacoustic flow cytography and on-the-spot laser killing of single circulating melanoma cells,” Sci. Rep., 6 (1), 39616 https://doi.org/10.1038/srep39616 (2016). Google Scholar

173. 

C. Kim et al., “In vivo molecular photoacoustic tomography of melanomas targeted by bioconjugated gold nanocages,” ACS Nano, 4 (8), 4559 –4564 https://doi.org/10.1021/nn100736c ANCAC3 1936-0851 (2010). Google Scholar

174. 

G. C. Langhout et al., “Detection of melanoma metastases in resected human lymph nodes by noninvasive multispectral photoacoustic imaging,” Int. J. Biomed. Imaging, 2014 163652 https://doi.org/10.1155/2014/163652 (2014). Google Scholar

175. 

X. Li et al., “A preliminary study of photoacoustic/ultrasound dual-mode imaging in melanoma using MAGE-targeted gold nanoparticles,” Biochem. Biophys. Res. Commun., 502 (2), 255 –261 https://doi.org/10.1016/j.bbrc.2018.05.155 (2018). Google Scholar

176. 

D. McCormack et al., “Photoacoustic detection of melanoma micrometastasis in sentinel lymph nodes,” J. Biomech. Eng., 131 (7), 074519 https://doi.org/10.1115/1.3169247 JBENDY 0148-0731 (2009). Google Scholar

177. 

J. Kim et al., “Multispectral ex vivo photoacoustic imaging of cutaneous melanoma for better selection of the excision margin,” Br. J. Dermatol., 179 (3), 780 –782 https://doi.org/10.1111/bjd.16677 BJDEAZ 0007-0963 (2018). Google Scholar

178. 

D. R. McCormack et al., “Enhanced photoacoustic detection of melanoma cells using gold nanoparticles,” Lasers Surg. Med., 43 (4), 333 –338 https://doi.org/10.1002/lsm.21060 LSMEDI 0196-8092 (2011). Google Scholar

179. 

Y. A. Menyaev et al., “Optical clearing in photoacoustic flow cytometry,” Biomed. Opt. Express, 4 (12), 3030 –3041 https://doi.org/10.1364/BOE.4.003030 BOEICL 2156-7085 (2013). Google Scholar

180. 

V. Neuschmelting et al., “Lymph node micrometastases and in-transit metastases from melanoma: in vivo detection with multispectral optoacoustic imaging in a mouse model,” Radiology, 280 (1), 137 –150 https://doi.org/10.1148/radiol.2016160191 RADLAX 0033-8419 (2016). Google Scholar

181. 

C. M. O’Brien et al., “Detection and isolation of circulating melanoma cells using photoacoustic flowmetry,” J. Vis. Exp., 57 e3559 https://doi.org/10.3791/3559 (2011). Google Scholar

182. 

M. Omar et al., “Pushing the optical imaging limits of cancer with multi-frequency-band raster-scan optoacoustic mesoscopy (RSOM),” Neoplasia, 17 (2), 208 –214 https://doi.org/10.1016/j.neo.2014.12.010 (2015). Google Scholar

183. 

A. J. Sinnamon et al., “Multispectral photoacoustic imaging for the detection of subclinical melanoma,” J. Surg. Oncol., 119 (8), 1070 –1076 https://doi.org/10.1002/jso.25447 JSONAU 0022-4790 (2019). Google Scholar

184. 

Y. Wang et al., “Toward in vivo biopsy of melanoma based on photoacoustic and ultrasound dual imaging with an integrated detector,” Biomed. Opt. Express, 7 (2), 279 –286 https://doi.org/10.1364/BOE.7.000279 BOEICL 2156-7085 (2016). Google Scholar

185. 

Q. Zhao et al., “Quantitative analysis on in vivo tumor‐microvascular images from optical-resolution photoacoustic microscopy,” J. Biophotonics, 12 (6), e201800421 https://doi.org/10.1002/jbio.201800421 (2019). Google Scholar

186. 

W. Zhou et al., “All-optical photoacoustic and reflectance confocal microscopy for melanoma characterization,” Appl. Phys. Lett., 114 (16), 163704 https://doi.org/10.1063/1.5087906 APPLAB 0003-6951 (2019). Google Scholar

187. 

Y. Zhou et al., “Handheld photoacoustic probe to detect both melanoma depth and volume at high speed in vivo,” J. Biophotonics, 8 (11–12), 961 –967 https://doi.org/10.1002/jbio.201400143 (2015). Google Scholar

188. 

Y. Zhou et al., “Handheld photoacoustic microscopy to detect melanoma depth in vivo,” Opt. Lett., 39 (16), 4731 –4734 https://doi.org/10.1364/OL.39.004731 OPLEDP 0146-9592 (2014). Google Scholar

189. 

G. He et al., “In vivo cell characteristic extraction and identification by photoacoustic flow cytography,” Biomed. Opt. Express, 6 (10), 3748 –3756 https://doi.org/10.1364/BOE.6.003748 BOEICL 2156-7085 (2015). Google Scholar

190. 

D. A. Nedosekin et al., “In vivo ultra-fast photoacoustic flow cytometry of circulating human melanoma cells using near-infrared high-pulse rate lasers,” Cytometry Part A, 79 (10), 825 –833 https://doi.org/10.1002/cyto.a.21102 1552-4922 (2011). Google Scholar

191. 

X. L. Deán-Ben et al., “Deep tissue volumetric optoacoustic tracking of individual circulating tumor cells in an intracardially perfused mouse model,” Neoplasia, 22 (9), 441 –446 https://doi.org/10.1016/j.neo.2020.06.008 (2020). Google Scholar

192. 

P. Hai et al., “Label-free high-throughput photoacoustic tomography of suspected circulating melanoma tumor cells in patients in vivo,” J. Biomed. Opt., 25 (3), 036002 https://doi.org/10.1117/1.JBO.25.3.036002 JBOPFO 1083-3668 (2020). Google Scholar

193. 

M. Han et al., “In vivo lymphatic circulating tumor cells and progression of metastatic disease,” Cancers, 12 (10), 2866 https://doi.org/10.3390/cancers12102866 (2020). Google Scholar

194. 

M. Hessler et al., “Melanoma biomarkers and their potential application for in vivo diagnostic imaging modalities,” Int. J. Mol. Sci., 21 (24), 9583 https://doi.org/10.3390/ijms21249583 1422-0067 (2020). Google Scholar

195. 

M. Wu et al., “Melanoma cell membrane biomimetic versatile CuS nanoprobes for homologous targeting photoacoustic imaging and photothermal chemotherapy,” ACS Appl. Mater. Interfaces, 12 (14), 16031 –16039 https://doi.org/10.1021/acsami.9b23177 (2020). Google Scholar

196. 

P. Armanetti et al., “Enhanced antitumoral activity and photoacoustic imaging properties of AuNP-enriched endothelial colony forming cells on melanoma,” Adv. Sci., 8 (4), 2001175 https://doi.org/10.1002/advs.202001175 1936-6612 (2021). Google Scholar

197. 

J. Kim et al., “Towards clinical photoacoustic and ultrasound imaging: probe improvement and real-time graphical user interface,” Exp. Biol. Med. (Maywood), 245 (4), 321 –329 https://doi.org/10.1177/1535370219889968 EXBMAA 0071-3384 (2020). Google Scholar

198. 

R. H. Edgar et al., “Capture and isolation of circulating melanoma cells using photoacoustic flowmetry,” Melanoma, 203 –212 Springer( (2021). Google Scholar

199. 

R. H. Edgar et al., “Predicting metastasis in melanoma by enumerating circulating tumor cells using photoacoustic flow cytometry,” Lasers Surg. Med., 53 (4), 578 –586 https://doi.org/10.1002/lsm.23286 LSMEDI 0196-8092 (2021). Google Scholar

200. 

R. M. Weight et al., “Photoacoustic detection of metastatic melanoma cells in the human circulatory system,” Opt. Lett., 31 (20), 2998 –3000 https://doi.org/10.1364/OL.31.002998 OPLEDP 0146-9592 (2006). Google Scholar

201. 

S. H. Holan and J. A. Viator, “Automated wavelet denoising of photoacoustic signals for circulating melanoma cell detection and burn image reconstruction,” Phys. Med. Biol., 53 (12), N227 https://doi.org/10.1088/0031-9155/53/12/N01 PHMBA7 0031-9155 (2008). Google Scholar

202. 

E. I. Galanzha et al., “In vivo fiber-based multicolor photoacoustic detection and photothermal purging of metastasis in sentinel lymph nodes targeted by nanoparticles,” J. Biophotonics, 2 528 –539 https://doi.org/10.1002/jbio.200910046 (2009). Google Scholar

203. 

G. Gutierrez-Juarez et al., “Detection of melanoma cells in vitro using an optical detector of photoacoustic waves,” Lasers Surg. Med., 42 (3), 274 –281 https://doi.org/10.1002/lsm.20894 LSMEDI 0196-8092 (2010). Google Scholar

204. 

Y. Zhang et al., “Chronic label-free volumetric photoacoustic microscopy of melanoma cells in three-dimensional porous scaffolds,” Biomaterials, 31 (33), 8651 –8658 https://doi.org/10.1016/j.biomaterials.2010.07.089 BIMADU 0142-9612 (2010). Google Scholar

205. 

C. Zhang, K. Maslov and L. V. Wang, “Subwavelength-resolution label-free photoacoustic microscopy of optical absorption in vivo,” Opt. Lett., 35 (19), 3195 –3197 https://doi.org/10.1364/OL.35.003195 OPLEDP 0146-9592 (2010). Google Scholar

206. 

J. Jose et al., “Initial results of imaging melanoma metastasis in resected human lymph nodes using photoacoustic computed tomography,” J. Biomed. Opt., 16 (9), 096021 https://doi.org/10.1117/1.3631705 JBOPFO 1083-3668 (2011). Google Scholar

207. 

C. M. O’Brien et al., “Capture of circulating tumor cells using photoacoustic flowmetry and two phase flow,” J. Biomed. Opt., 17 (6), 061221 https://doi.org/10.1117/1.JBO.17.6.061221 JBOPFO 1083-3668 (2012). Google Scholar

208. 

S. Gupta et al., “Photoacoustic detection of induced melanoma in vitro using a mouse model,” Proc. SPIE, 8214 821414 https://doi.org/10.1117/12.909352 PSISDG 0277-786X (2012). Google Scholar

209. 

Y. Yang et al., “Photoacoustic tomography of tissue subwavelength microstructure with a narrowband and low frequency system,” Appl. Phys. Lett., 101 (3), 034105 https://doi.org/10.1063/1.4736994 APPLAB 0003-6951 (2012). Google Scholar

210. 

C. Zhang et al., “In vivo photoacoustic microscopy with 7.6-μm axial resolution using a commercial 125-MHz ultrasonic transducer,” J. Biomed. Opt., 17 (11), 116016 https://doi.org/10.1117/1.JBO.17.11.116016 JBOPFO 1083-3668 (2012). Google Scholar

211. 

D. A. Nedosekin et al., “Synergy of photoacoustic and fluorescence flow cytometry of circulating cells with negative and positive contrasts,” J. Biophotonics, 6 (5), 425 –434 https://doi.org/10.1002/jbio.201200047 (2013). Google Scholar

212. 

D. A. Nedosekin et al., “Photoacoustic and photothermal detection of circulating tumor cells, bacteria and nanoparticles in cerebrospinal fluid in vivo and ex vivo,” J. Biophotonics, 6 523 –533 https://doi.org/10.1002/jbio.201200242 (2013). Google Scholar

213. 

M. Sarimollaoglu et al., “Nonlinear photoacoustic signal amplification from single targets in absorption background,” Photoacoustics, 2 (1), 1 –11 https://doi.org/10.1016/j.pacs.2013.11.002 (2014). Google Scholar

214. 

L. Xiang et al., “Label-free photoacoustic cell-tracking in real-time,” X Acoust. Imaging Sens., 1 18 –22 https://doi.org/10.2478/phto-2014-0002 (2014). Google Scholar

215. 

M. A. Juratli et al., “Real-time monitoring of circulating tumor cell release during tumor manipulation using in vivo photoacoustic and fluorescent flow cytometry,” Head Neck, 36 (8), 1207 –1215 https://doi.org/10.1002/hed.23439 (2014). Google Scholar

216. 

J. Yao et al., “Photoimprint photoacoustic microscopy for three-dimensional label-free subdiffraction imaging,” Phys. Rev. Lett., 112 (1), 014302 https://doi.org/10.1103/PhysRevLett.112.014302 PRLTAO 0031-9007 (2014). Google Scholar

217. 

W. Song et al., “Reflection-mode in vivo photoacoustic microscopy with subwavelength lateral resolution,” Biomed. Opt. Express, 5 (12), 4235 –4241 https://doi.org/10.1364/BOE.5.004235 BOEICL 2156-7085 (2014). Google Scholar

218. 

A. Breathnach et al., “Assessment of cutaneous melanoma and pigmented skin lesions with photoacoustic imaging,” Proc. SPIE, 9303 930303 https://doi.org/10.1117/12.2078309 PSISDG 0277-786X (2015). Google Scholar

219. 

M.-J. Sun and P.-C. Li, “Imaging 3D cell culture systems using an optical resolution photoacoustic microscope,” in IEEE Int. Ultrasonics Symp., (2014). https://doi.org/10.1109/ULTSYM.2014.0312 Google Scholar

220. 

C. Lee et al., “Real-time near-infrared virtual intraoperative surgical photoacoustic microscopy,” Photoacoustics, 3 (3), 100 –106 https://doi.org/10.1016/j.pacs.2015.08.002 (2015). Google Scholar

221. 

T. Wang et al., “All-optical photoacoustic microscopy based on plasmonic detection of broadband ultrasound,” Appl. Phys. Lett., 107 (15), 153702 https://doi.org/10.1063/1.4933333 APPLAB 0003-6951 (2015). Google Scholar

222. 

Y. Zhou et al., “In vivo melanoma depth detection by a handheld photoacoustic microscope,” Proc. SPIE, 9323 932304 https://doi.org/10.1117/12.2076684 PSISDG 0277-786X (2015). Google Scholar

223. 

K. Bhattacharyya et al., “Quantitative photoacoustics to measure single cell melanin production and nanoparticle attachment,” Phys. Med. Biol., 60 (8), 3081 https://doi.org/10.1088/0031-9155/60/8/3081 PHMBA7 0031-9155 (2015). Google Scholar

224. 

Y. Zhou et al., “Detecting both melanoma depth and volume in vivo with a handheld photoacoustic probe,” Proc. SPIE, 9708 970806 https://doi.org/10.1117/12.2213245 PSISDG 0277-786X (2016). Google Scholar

225. 

M. Neuwirth et al., “Detection of melanoma metastases in regional lymph nodes using multispectral photoacoustic imaging,” J. Am. Coll. Surg., 225 (4), S189 https://doi.org/10.1016/j.jamcollsurg.2017.07.431 JACSEX 1072-7515 (2017). Google Scholar

226. 

K. Sivasubramanian et al., “Near-infrared light-responsive liposomal contrast agent for photoacoustic imaging and drug release applications,” J. Biomed. Opt., 22 (4), 041007 https://doi.org/10.1117/1.JBO.22.4.041007 JBOPFO 1083-3668 (2017). Google Scholar

227. 

P. Hajireza et al., “Non-interferometric photoacoustic remote sensing microscopy,” Light: Sci. Appl., 6 (6), e16278 https://doi.org/10.1038/lsa.2016.278 (2017). Google Scholar

228. 

C. Liu et al., “Switchable photoacoustic imaging of glutathione using MnO2 nanotubes for cancer diagnosis,” ACS Appl. Mater. Interfaces, 10 (51), 44231 –44239 https://doi.org/10.1021/acsami.8b14944 (2018). Google Scholar

229. 

Q. Zhou et al., “Label-free counting of circulating cells by in vivo photoacoustic flow cytometry,” Proc. SPIE, 10495 104950T https://doi.org/10.1117/12.2289216 PSISDG 0277-786X (2018). Google Scholar

230. 

Q. Zhou et al., “In vivo and label-free detection of circulating melanoma cells by photoacoustic flow cytometry,” in Conf. Lasers and Electro-Opt./Pac. Rim, (2018). Google Scholar

231. 

H. Chen et al., “Optical-resolution photoacoustic microscopy using transparent ultrasound transducer,” Sensors, 19 (24), 5470 https://doi.org/10.3390/s19245470 SNSRES 0746-9462 (2019). Google Scholar

232. 

W. Zhou et al., “Optical biopsy of melanoma and basal cell carcinoma progression by noncontact photoacoustic and optical coherence tomography: in vivo multi-parametric characterizing tumor microenvironment,” IEEE Trans. Med. Imaging, 39 (6), 1967 –1974 https://doi.org/10.1109/TMI.2019.2962614 ITMID4 0278-0062 (2019). Google Scholar

233. 

Y. Chang et al., “Co-impulse multispectral photoacoustic microscopy and optical coherence tomography system using a single supercontinuum laser,” Opt. Lett., 44 (18), 4459 –4462 https://doi.org/10.1364/OL.44.004459 OPLEDP 0146-9592 (2019). Google Scholar

234. 

N. Liu, Z. Chen and D. Xing, “Integrated photoacoustic and hyperspectral dual-modality microscopy for co-imaging of melanoma and cutaneous squamous cell carcinoma in vivo,” J. Biophotonics, 13 (8), e202000105 https://doi.org/10.1002/jbio.202000105 (2020). Google Scholar

235. 

Q. Liu et al., “Label-free counting of circulating melanoma cells in deep vessels with photoacoustic flow cytometry,” Proc. SPIE, 11241 112410T https://doi.org/10.1117/12.2543931 PSISDG 0277-786X (2020). Google Scholar

236. 

R. Shintate et al., “Development of optical resolution photoacoustic microscopy with sub-micron lateral resolution for visualization of cells and their structures,” Jpn. J. Appl. Phys., 59 (SK), SKKE11 https://doi.org/10.35848/1347-4065/ab840e JJAPA5 0021-4922 (2020). Google Scholar

237. 

L. Pires et al., “Dual-agent photodynamic therapy with optical clearing eradicates pigmented melanoma in preclinical tumor models,” Cancers, 12 (7), 1956 https://doi.org/10.3390/cancers12071956 (2020). Google Scholar

238. 

Z. Wang et al., “Photoacoustic and ultrasound (PAUS) dermoscope with high sensitivity and penetration depth by using a bimorph transducer,” J. Biophotonics, 13 (9), e202000145 https://doi.org/10.1002/jbio.202000145 (2020). Google Scholar

239. 

B. Park et al., “A photoacoustic finder fully integrated with a solid-state dye laser and transparent ultrasound transducer,” Photoacoustics, 23 100290 https://doi.org/10.1016/j.pacs.2021.100290 (2021). Google Scholar

240. 

T. T. Mai et al., “In vivo quantitative vasculature segmentation and assessment for photodynamic therapy process monitoring using photoacoustic microscopy,” Sensors, 21 (5), 1776 https://doi.org/10.3390/s21051776 SNSRES 0746-9462 (2021). Google Scholar

241. 

J. Park et al., “Quadruple ultrasound, photoacoustic, optical coherence, and fluorescence fusion imaging with a transparent ultrasound transducer,” Proc. Natl. Acad. Sci. U. S. A., 118 (11), e1920879118 https://doi.org/10.1073/pnas.1920879118 (2021). Google Scholar

242. 

M. T. Stridh et al., “Photoacoustic imaging of periorbital skin cancer ex vivo: unique spectral signatures of malignant melanoma, basal, and squamous cell carcinoma,” Biomed. Opt. Express, 13 (1), 410 –425 https://doi.org/10.1364/BOE.443699 BOEICL 2156-7085 (2022). Google Scholar

243. 

H. Ma et al., “Multiscale confocal photoacoustic dermoscopy to evaluate skin health,” Quantum Imaging Med. Surg., 12 (5), 2696 https://doi.org/10.21037/qims-21-878 (2022). Google Scholar

244. 

A. F. Kukk et al., “Multimodal imaging system with ultrasound, photoacoustics, and optical coherence tomography for optical biopsy of melanoma,” Proc. SPIE, 12371 1237107 https://doi.org/10.1117/12.2648257 PSISDG 0277-786X (2023). Google Scholar

245. 

P. M. Patil et al., “Optoresponsive pheophorbide-silver based organometallic nanomaterials for high efficacy multimodal theranostics in Melanoma,” Chem. Eng. J., 470 144110 https://doi.org/10.1016/j.cej.2023.144110 (2023). Google Scholar

246. 

D. J. Karimipour et al., “Microstaging accuracy after subtotal incisional biopsy of cutaneous melanoma,” J. Am. Acad. Dermatol., 52 (5), 798 –802 https://doi.org/10.1016/j.jaad.2004.09.031 JAADDB 0190-9622 (2005). Google Scholar

247. 

J. K. Mills et al., “Effect of biopsy type on outcomes in the treatment of primary cutaneous melanoma,” Am. J. Surg., 205 (5), 585 –590 https://doi.org/10.1016/j.amjsurg.2013.01.023 AJOOA7 0096-6347 (2013). Google Scholar

248. 

T. J. Hieken et al., “Accuracy of diagnostic biopsy for cutaneous melanoma: implications for surgical oncologists,” Int. J. Surg. Oncol., 2013 196493 https://doi.org/10.1155/2013/196493 (2013). Google Scholar

249. 

P. Moore et al., “Does shave biopsy accurately predict the final Breslow depth of primary cutaneous melanoma?,” Am. Surg., 75 (5), 369 –373 https://doi.org/10.1177/000313480907500503 (2009). Google Scholar

250. 

J. S. Zager et al., “Shave biopsy is a safe and accurate method for the initial evaluation of melanoma,” J. Am. Coll. Surg., 212 (4), 454 –460 https://doi.org/10.1016/j.jamcollsurg.2010.12.021 JACSEX 1072-7515 (2011). Google Scholar

251. 

C. Wang et al., “In-vivo imaging of melanoma with simultaneous dual-wavelength acoustic-resolution-based photoacoustic/ultrasound microscopy,” Appl. Opt., 60 (13), 3772 –3778 https://doi.org/10.1364/AO.412609 APOPAI 0003-6935 (2021). Google Scholar

252. 

J. A. Swearingen et al., “Photoacoustic discrimination of vascular and pigmented lesions using classical and Bayesian methods,” J. Biomed. Opt., 15 (1), 016019 https://doi.org/10.1117/1.3316297 JBOPFO 1083-3668 (2010). Google Scholar

253. 

N. Lassau et al., “Prognostic value of angiogenesis evaluated with high-frequency and colour Doppler sonography for preoperative assessment of primary cutaneous melanomas: correlation with recurrence after a 5 year follow-up period,” Cancer Imaging, 6 (1), 24 https://doi.org/10.1102/1470-7330.2006.0009 (2006). Google Scholar

254. 

G. Ku et al., “Imaging of tumor angiogenesis in rat brains in vivo by photoacoustic tomography,” Appl. Opt., 44 (5), 770 –775 https://doi.org/10.1364/AO.44.000770 APOPAI 0003-6935 (2005). Google Scholar

255. 

A. Breslow, “Thickness, cross-sectional areas and depth of invasion in the prognosis of cutaneous melanoma,” Ann. Surg., 172 (5), 902 –908 https://doi.org/10.1097/00000658-197011000-00017 (1970). Google Scholar

256. 

G. Manca et al., “Sentinel lymph node mapping in melanoma: the issue of false-negative findings,” Clin. Nucl. Med., 39 (7), e346 –e354 https://doi.org/10.1097/RLU.0000000000000366 CNMEDK 0363-9762 (2014). Google Scholar

257. 

S. P. Leong, S.-R. Kothapalli and S. S. Gambhir, “Can multispectral optoacoustic tomography replace sentinel lymph biopsy in melanoma?,” Ann. Transl. Med., 4 (24), 517 https://doi.org/10.21037/atm.2016.12.31 (2016). Google Scholar

258. 

I. Stoffels et al., “Assessment of nonradioactive multispectral optoacoustic tomographic imaging with conventional lymphoscintigraphic imaging for sentinel lymph node biopsy in melanoma,” JAMA Network Open, 2 (8), e199020 https://doi.org/10.1001/jamanetworkopen.2019.9020 (2019). Google Scholar

259. 

L. M. De Souza, B. M. Robertson and G. P. Robertson, “Future of circulating tumor cells in the melanoma clinical and research laboratory settings,” Cancer Lett., 392 60 –70 https://doi.org/10.1016/j.canlet.2017.01.023 CALEDQ 0304-3835 (2017). Google Scholar

260. 

M. Liu et al., “Circulating tumor cells: a new window for diagnosis and evaluation of cancer,” Anti-Cancer Agents Med. Chem. (Formerly Curr. Med. Chem.-Anti-Cancer Agents), 16 (12), 1529 –1540 https://doi.org/10.2174/1871520616666160223113743 (2016). Google Scholar

261. 

T. Bauernhofer et al., “Association of disease progression and poor overall survival with detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer,” Oncol. Rep., 13 (2), 179 –184 https://doi.org/10.3892/or.13.2.179 OCRPEW 1021-335X (2005). Google Scholar

262. 

M. Cristofanilli et al., “Circulating tumor cells, disease progression, and survival in metastatic breast cancer,” N. Engl. J. Med., 351 (8), 781 –791 https://doi.org/10.1056/NEJMoa040766 NEJMBH (2004). Google Scholar

263. 

M. C. Miller, G. V. Doyle and L. W. Terstappen, “Significance of circulating tumor cells detected by the CellSearch system in patients with metastatic breast colorectal and prostate cancer,” J. Oncol., 2010 617421 https://doi.org/10.1155/2010/617421 (2010). Google Scholar

264. 

E. I. Galanzha and V. P. Zharov, “Photoacoustic flow cytometry,” Methods, 57 (3), 280 –296 https://doi.org/10.1016/j.ymeth.2012.06.009 MTHDE9 1046-2023 (2012). Google Scholar

265. 

E. I. Galanzha and V. P. Zharov, “Circulating tumor cell detection and capture by photoacoustic flow cytometry in vivo and ex vivo,” Cancers, 5 (4), 1691 –1738 https://doi.org/10.3390/cancers5041691 (2013). Google Scholar

266. 

M. Yu et al., “Circulating tumor cells: approaches to isolation and characterization,” J. Cell Biol., 192 (3), 373 –382 https://doi.org/10.1083/jcb.201010021 JCLBA3 0021-9525 (2011). Google Scholar

267. 

S. Abbasi et al., “All-optical reflection-mode microscopic histology of unstained human tissues,” Sci. Rep., 9 (1), 13392 https://doi.org/10.1038/s41598-019-49849-9 (2019). Google Scholar

268. 

K. Bell et al., “Reflection-mode virtual histology using photoacoustic remote sensing microscopy,” Sci. Rep., 10 (1), 19121 https://doi.org/10.1038/s41598-020-76155-6 (2020). Google Scholar

269. 

B. Ecclestone et al., “Label-free histological imaging of skin cancers with photoacoustic remote sensing virtual histology,” Proc. SPIE, PC11934 PC119340F https://doi.org/10.1117/12.2609176 PSISDG 0277-786X (2022). Google Scholar

270. 

R. E. Benjamin et al., “Exploration of photoacoustic remote sensing virtual histology in skin malignancies,” Proc. SPIE, PC12352 PC1235201 https://doi.org/10.1117/12.2650980 PSISDG 0277-786X (2023). Google Scholar

271. 

J. Weber, P. C. Beard and S. E. Bohndiek, “Contrast agents for molecular photoacoustic imaging,” Nat. Methods, 13 (8), 639 –650 https://doi.org/10.1038/nmeth.3929 1548-7091 (2016). Google Scholar

272. 

V. P. Zharov et al., “In vivo photoacoustic flow cytometry for monitoring of circulating single cancer cells and contrast agents,” Opt. Lett., 31 (24), 3623 –3625 https://doi.org/10.1364/OL.31.003623 OPLEDP 0146-9592 (2006). Google Scholar

273. 

W. Li et al., “Gold nanocages as contrast agents for photoacoustic imaging,” Contrast Media Mol. Imaging, 6 (5), 370 –377 https://doi.org/10.1002/cmmi.439 (2011). Google Scholar

274. 

K. Bhattacharyya et al., “Gold nanoparticle–mediated detection of circulating cancer cells,” Clin. Lab. Med., 32 (1), 89 –101 https://doi.org/10.1016/j.cll.2012.01.001 CLMED6 0272-2712 (2012). Google Scholar

275. 

D. Chenthamara et al., “Therapeutic efficacy of nanoparticles and routes of administration,” Biomater. Res., 23 20 https://doi.org/10.1186/s40824-019-0166-x (2019). Google Scholar

276. 

A. Onaciu et al., “Theranostic nanoparticles and their spectrum in cancer,” Engineered Nanomaterials-Health and Safety, IntechOpen( (2019). Google Scholar

277. 

H. H. Gustafson et al., “Nanoparticle uptake: the phagocyte problem,” Nano Today, 10 (4), 487 –510 https://doi.org/10.1016/j.nantod.2015.06.006 NTAOCG 1748-0132 (2015). Google Scholar

278. 

A. J. Tavares et al., “Effect of removing Kupffer cells on nanoparticle tumor delivery,” Proc. Natl. Acad. Sci. U. S. A., 114 (51), E10871 –E10880 https://doi.org/10.1073/pnas.1713390114 (2017). Google Scholar

279. 

Q. Fu et al., “Photoacoustic imaging: contrast agents and their biomedical applications,” Adv. Mater., 31 (6), 1805875 https://doi.org/10.1002/adma.201805875 ADVMEW 0935-9648 (2019). Google Scholar

280. 

A. Farooq et al., “Exogenous contrast agents in photoacoustic imaging: an in vivo review for tumor imaging,” Nanomaterials, 12 (3), 393 https://doi.org/10.3390/nano12030393 (2022). Google Scholar

281. 

R. R. Braeuer et al., “Galectin-3 contributes to melanoma growth and metastasis via regulation of NFAT1 and autotaxin,” Cancer Res., 72 (22), 5757 –5766 https://doi.org/10.1158/0008-5472.CAN-12-2424 CNREA8 0008-5472 (2012). Google Scholar

282. 

H. Zhong et al., “Review of low-cost photoacoustic sensing and imaging based on laser diode and light-emitting diode,” Sensors, 18 (7), 2264 https://doi.org/10.3390/s18072264 SNSRES 0746-9462 (2018). Google Scholar

283. 

A. Fatima et al., “Review of cost reduction methods in photoacoustic computed tomography,” Photoacoustics, 15 100137 https://doi.org/10.1016/j.pacs.2019.100137 (2019). Google Scholar

284. 

M. Zafar et al., “Low-cost fast photoacoustic computed tomography: phantom study,” Proc. SPIE, 10878 108785V https://doi.org/10.1117/12.2510552 PSISDG 0277-786X (2019). Google Scholar

285. 

Q. Chen et al., “Progress of clinical translation of handheld and semi-handheld photoacoustic imaging,” Photoacoustics, 22 100264 https://doi.org/10.1016/j.pacs.2021.100264 (2021). Google Scholar

286. 

J. Lee et al., “Design and implementation of embedded-based vein image processing system with enhanced denoising capabilities,” Sensors, 22 (21), 8559 https://doi.org/10.3390/s22218559 SNSRES 0746-9462 (2022). Google Scholar

287. 

A. B. E. Attia et al., “Microvascular imaging and monitoring of hemodynamic changes in the skin during arterial-venous occlusion using multispectral raster-scanning optoacoustic mesoscopy,” Photoacoustics, 22 100268 https://doi.org/10.1016/j.pacs.2021.100268 (2021). Google Scholar

288. 

M. Moothanchery et al., “Monitoring skin microvascular hemodynamic changes using multispectral optoacoustic mesoscopy,” in Bio-Opt.: Des. and Appl., (2021). Google Scholar

289. 

X. Li et al., “Multispectral raster-scanning optoacoustic mesoscopy differentiate lesional from non-lesional atopic dermatitis skin using structural and functional imaging markers,” Photoacoustics, 28 100399 https://doi.org/10.1016/j.pacs.2022.100399 (2022). Google Scholar

290. 

M. V. Novoselova et al., “Optical clearing for photoacoustic lympho-and angiography beyond conventional depth limit in vivo,” Photoacoustics, 20 100186 https://doi.org/10.1016/j.pacs.2020.100186 (2020). Google Scholar

291. 

L. Hacker et al., “Performance evaluation of mesoscopic photoacoustic imaging,” Photoacoustics, 31 100505 https://doi.org/10.1016/j.pacs.2023.100505 (2023). Google Scholar

292. 

J. Aguirre et al., “Precision assessment of label-free psoriasis biomarkers with ultra-broadband optoacoustic mesoscopy,” Nat. Biomed. Eng., 1 (5), 0068 https://doi.org/10.1038/s41551-017-0068 (2017). Google Scholar

293. 

Y. W. Yew et al., “Raster-scanning optoacoustic mesoscopy imaging as an objective disease severity tool in atopic dermatitis patients,” J. Am. Acad. Dermatol., 84 (4), 1121 –1123 https://doi.org/10.1016/j.jaad.2020.06.045 JAADDB 0190-9622 (2021). Google Scholar

294. 

H. Zhou et al., “Exploring a fluorescent blood pool agent in photoacoustic imaging,” J. Nucl. Med., 57 1214 JNMEAQ 0161-5505 (2016). Google Scholar

295. 

W. M. MacCuaig et al., “Development of multispectral optoacoustic tomography as a clinically translatable modality for cancer imaging,” Radiol.: Imaging Cancer, 2 (6), e200066 https://doi.org/10.1148/rycan.2020200066 CANCAR 0008-543X (2020). Google Scholar

296. 

L. Monteiro Rodrigues, T. F. Granja and S. F. de Andrade, “Optoacoustic imaging offers new insights into in vivo human skin vascular physiology,” Life, 12 (10), 1628 https://doi.org/10.3390/life12101628 (2022). Google Scholar

297. 

K. S. Valluru and J. K. Willmann, “Clinical photoacoustic imaging of cancer,” Ultrasonography, 35 (4), 267 https://doi.org/10.14366/usg.16035 (2016). Google Scholar

298. 

H. Zafar et al., “Photoacoustic imaging of the human forearm using 40 MHz linear-array transducer,” Proc. SPIE, 8942 89420M https://doi.org/10.1117/12.2040637 PSISDG 0277-786X (2014). Google Scholar

299. 

Z. Sun et al., “TiL4 -coordinated black phosphorus quantum dots as an efficient contrast agent for in vivo photoacoustic imaging of cancer,” Small, 13 1602896 https://doi.org/10.1002/smll.201602896 SMALBC 1613-6810 (2017). Google Scholar

300. 

C. Zhang et al., “Photoacoustic and fluorescence imaging of cutaneous squamous cell carcinoma in living subjects using a probe targeting integrin αvβ6,” Sci. Rep., 7 (1), 42442 https://doi.org/10.1038/srep42442 (2017). Google Scholar

301. 

D. Van de Sompel et al., “Improving image quality by accounting for changes in water temperature during a photoacoustic tomography scan,” PLoS One, 7 (10), e45337 https://doi.org/10.1371/journal.pone.0045337 (2012). Google Scholar

302. 

J. V. Jokerst et al., “Gold nanorods for ovarian cancer detection with photoacoustic imaging and resection guidance via Raman imaging in living mice,” ACS Nano, 6 (11), 10366 –10377 https://doi.org/10.1021/nn304347g ANCAC3 1936-0851 (2012). Google Scholar

303. 

Y. Liu et al., “Porous gold nanocluster-decorated manganese monoxide nanocomposites for microenvironment-activatable MR/photoacoustic/CT tumor imaging,” Nanoscale, 10 (8), 3631 –3638 https://doi.org/10.1039/C7NR08535D NANOHL 2040-3364 (2018). Google Scholar

304. 

A. Liopo, R. Su and A. A. Oraevsky, “Melanin nanoparticles as a novel contrast agent for optoacoustic tomography,” Photoacoustics, 3 (1), 35 –43 https://doi.org/10.1016/j.pacs.2015.02.001 (2015). Google Scholar

305. 

A. Oraevsky, “Three-dimensional quantitative optoacoustic tomography,” in Int. Conf. Photoacoustic and Photothermal Phenomena, (2022). Google Scholar

306. 

A. Liopo et al., “Optical clearing of skin enhanced with hyaluronic acid for increased contrast of optoacoustic imaging,” J. Biomed. Opt., 21 (8), 081208 https://doi.org/10.1117/1.JBO.21.8.081208 JBOPFO 1083-3668 (2016). Google Scholar

307. 

S. A. Ermilov et al., “3D laser optoacoustic ultrasonic imaging system for research in mice (LOUIS-3DM),” Proc. SPIE, 8943 89430J https://doi.org/10.1117/12.2044817 PSISDG 0277-786X (2014). Google Scholar

308. 

J. Yao and N. Van Tu, “High-speed photoacoustic microscopy captures processes within tissue,” Google Scholar

309. 

M. Kuniyil Ajith Singh et al., “Clinical translation of photoacoustic imaging—opportunities and challenges from an industry perspective,” LED-Based Photoacoustic Imaging: From Bench to Bedside, 379 –393 Springer( (2020). Google Scholar

310. 

H.-Z. Gong, H.-Y. Zheng and J. Li, “Amelanotic melanoma,” Melanoma Res., 29 (3), 221 –230 https://doi.org/10.1097/CMR.0000000000000571 MREEEH 0960-8931 (2019). Google Scholar

Biographies of the authors are not available.

CC BY: © The Authors. Published by SPIE under a Creative Commons Attribution 4.0 International License. Distribution or reproduction of this work in whole or in part requires full attribution of the original publication, including its DOI.
Joseph W. Fakhoury, Juliana Benavides Lara, Rayyan Manwar, Mohsin Zafar, Qiuyun Xu, Ricardo Engel, Maria M. Tsoukas, Steven Daveluy, Darius Mehregan, and Kamran Avanaki "Photoacoustic imaging for cutaneous melanoma assessment: a comprehensive review," Journal of Biomedical Optics 29(S1), S11518 (12 January 2024). https://doi.org/10.1117/1.JBO.29.S1.S11518
Received: 1 October 2023; Accepted: 21 December 2023; Published: 12 January 2024
Advertisement
Advertisement
KEYWORDS
Melanoma

Cancer detection

Tumors

Lymph nodes

Photoacoustic imaging

In vivo imaging

Acquisition tracking and pointing

Back to Top