Intraoperative fluorescence imaging with aminolevulinic acid detects grossly occult breast cancer: a phase II randomized controlled trial

Kathryn Ottolino-Perry, Anam Shahid, Stephanie DeLuca, Viktor Son, Mayleen Sukhram, Fannong Meng, Zhihui Amy Liu, Sara Rapic, Nayana Thalanki Anantha, Shirley C Wang, Emilie Chamma, Christopher Gibson, Philip J Medeiros, Safa Majeed, Ashley Chu, Olivia Wignall, Alessandra Pizzolato, Cheryl F Rosen, Liis Lindvere Teene, Danielle Starr-Dunham, Iris Kulbatski, Tony Panzarella, Susan J Done, Alexandra M Easson, Wey L Leong, Ralph S DaCosta, Kathryn Ottolino-Perry, Anam Shahid, Stephanie DeLuca, Viktor Son, Mayleen Sukhram, Fannong Meng, Zhihui Amy Liu, Sara Rapic, Nayana Thalanki Anantha, Shirley C Wang, Emilie Chamma, Christopher Gibson, Philip J Medeiros, Safa Majeed, Ashley Chu, Olivia Wignall, Alessandra Pizzolato, Cheryl F Rosen, Liis Lindvere Teene, Danielle Starr-Dunham, Iris Kulbatski, Tony Panzarella, Susan J Done, Alexandra M Easson, Wey L Leong, Ralph S DaCosta

Abstract

Background: Re-excision due to positive margins following breast-conserving surgery (BCS) negatively affects patient outcomes and healthcare costs. The inability to visualize margin involvement is a significant challenge in BCS. 5-Aminolevulinic acid hydrochloride (5-ALA HCl), a non-fluorescent oral prodrug, causes intracellular accumulation of fluorescent porphyrins in cancer cells. This single-center Phase II randomized controlled trial evaluated the safety, feasibility, and diagnostic accuracy of a prototype handheld fluorescence imaging device plus 5-ALA for intraoperative visualization of invasive breast carcinomas during BCS.

Methods: Fifty-four patients were enrolled and randomized to receive no 5-ALA or oral 5-ALA HCl (15 or 30 mg/kg). Forty-five patients (n = 15/group) were included in the analysis. Fluorescence imaging of the excised surgical specimen was performed, and biopsies were collected from within and outside the clinically demarcated tumor border of the gross specimen for blinded histopathology.

Results: In the absence of 5-ALA, tissue autofluorescence imaging lacked tumor-specific fluorescent contrast. Both 5-ALA doses caused bright red tumor fluorescence, with improved visualization of tumor contrasted against normal tissue autofluorescence. In the 15 mg/kg 5-ALA group, the positive predictive value (PPV) for detecting breast cancer inside and outside the grossly demarcated tumor border was 100.0% and 55.6%, respectively. In the 30 mg/kg 5-ALA group, the PPV was 100.0% and 50.0% inside and outside the demarcated tumor border, respectively. No adverse events were observed, and clinical feasibility of this imaging device-5-ALA combination approach was confirmed.

Conclusions: This is the first known clinical report of visualization of 5-ALA-induced fluorescence in invasive breast carcinoma using a real-time handheld intraoperative fluorescence imaging device.

Trial registration: Clinicaltrials.gov identifier NCT01837225 . Registered 23 April 2013.

Keywords: Aminolevulinic acid; Breast cancer; Breast-conserving surgery; Fluorescence imaging; Handheld intraoperative imaging device; Intraoperative imaging; Margin assessment; Optical imaging.

Conflict of interest statement

RSD, KOP, CG, NTA, WL, AE, and SD are co-inventors on IP licensed to MolecuLight Inc. (Toronto, Canada) and photonamic GmbH & Co. KG (Pinneberg, Germany) commercializing this technology. This study was a University Health Network (UHN) investigator-sponsored RCT (clinicaltrials.gov identifier NCT01837225) prior to IP licensing. RSD is the Founder, Chief Scientific Officer, Board member, and Shareholder of MolecuLight Inc. MolecuLight Inc. did not provide funding or other support, nor were any company staff involved directly or indirectly with this study. RSD conceived of the study and reviewed and edited the final manuscript. He did not recruit nor consent patients, nor collect, nor analyze data resulting from the study. 5-ALA HCl was provided by photonamic GmbH and Co. KG (Pinneberg, Germany) governed by a Drug Transfer Agreement between photonamic and UHN. RSD holds interest in photonamic GmbH & Co. KG’s subsidiary company (SBI ALApharma Canada Inc., Toronto, Canada) developing this technology for commercialization. KOP is employed by SBI ALApharma Canada Inc., but was solely employed by UHN during study data collection, data analysis and manuscript preparation. SBI ALApharma Canada Inc. was not involved directly or indirectly in this study. RSD is governed by a UHN Conflict of Interest Management Plan. All other authors declare that they have no competing interests.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
CONSORT diagram. Patients with tumors measuring

Fig. 2

Fluorescence imaging of 5-ALA-induced PpIX…

Fig. 2

Fluorescence imaging of 5-ALA-induced PpIX fluorescence in grossly obvious and grossly occult carcinoma.…

Fig. 2
Fluorescence imaging of 5-ALA-induced PpIX fluorescence in grossly obvious and grossly occult carcinoma. A Representative white light (top row) and fluorescence (bottom row) images of grossly obvious disease in sectioned lumpectomy specimens (a) and a clinically positive sentinel lymph node (c). The pathologist’s assistant (V.S., M.S., F.M.) demarcated tumor border (blue line) identified the grossly obvious tumor in the sectioned specimens. The surgeon (A.M.E., W.L.L.) identified the lymph node as grossly obvious for disease. B Representative white light (top row) and fluorescence (bottom row) images of grossly occult disease at the surface of an excised lumpectomy (a), in grossly sectioned specimens (b, c) and a sentinel lymph node (d) from patients with invasive ductal carcinoma with (a, d) or without (b, c) a DCIS component administered 15 mg/kg (b, c) or 30 mg/kg (a, d) 5-ALA HCl. Images represent tissue that was identified by the surgeon (A.M.E., W.L.L.) (a, d) or pathologist’s assistant (V.S., M.S., F.M.) (b, c) as grossly negative for the presence of cancer. (a) DCIS identified by fluorescence imaging at the lumpectomy margin. (b, c) Invasive carcinoma identified by fluorescence imaging on slices outside the grossly demarcated tumor. (d) Invasive carcinoma macro-metastases identified by fluorescence imaging in an excised sentinel lymph node. Scale bars = 5 mm

Fig. 3

Detection of grossly occult sub-millimeter…

Fig. 3

Detection of grossly occult sub-millimeter red fluorescence tumor foci. A WL and fluorescence…

Fig. 3
Detection of grossly occult sub-millimeter red fluorescence tumor foci. A WL and fluorescence images of a slice containing no clinically obvious disease from a patient who received 30 mg/kg 5-ALA HCl. Biopsies were collected in an area of focal red PpIX fluorescence (Bx1) and an adjacent area lacking PpIX fluorescence (Bx2). B H&E-stained longitudinal section of the Bx1 biopsy identified in A, which was determined to contain invasive ductal carcinoma by a blinded pathologist (S.J.D.). The imaged surface of the biopsy is indicated by the arrowheads. The area of tumor near the imaged surface measured 0.71 mm2. C H&E-stained longitudinal section of the Bx2 biopsy identified in A, which was determined to be negative for tumor by a blinded pathologist (S.J.D.). D White light and fluorescence images of slices of a lumpectomy from a patient who received 15 mg/kg 5-ALA HCl. A biopsy (Bx1) was collected from a small area of red fluorescence (inset digitally zoomed). E H&E-stained longitudinal section of the Bx1 identified in D, which was determined to contain DCIS > 2 mm below the imaged surface (arrowheads). Scale bar = 0.5 mm (A, D), 500 μm (B, C, E). WL, white light; FL, fluorescence; Bx, biopsy; IDC, invasive ductal carcinoma; DCIS, ductal carcinoma in situ

Fig. 4

Cancer cell-specific localization of ALA-induced…

Fig. 4

Cancer cell-specific localization of ALA-induced PpIX fluorescence. Representative A fluorescence microscopy and B…

Fig. 4
Cancer cell-specific localization of ALA-induced PpIX fluorescence. Representative A fluorescence microscopy and B corresponding histological images (bottom panel) from a biopsy collected inside the demarcated primary tumor boundary of a patient who received 30 mg/kg 5-ALA. The biopsy appeared red fluorescent with PRODIGI imaging. A Fluorescence microscopy was performed on cryosections cut from tumor core biopsies followed by B H&E, Masson’s Trichrome (MT), and Oil Red O (ORO) staining. Arrowheads depict green AF that was consistently observed in fibrous collagen tissue and in locations of necrosis, as confirmed with H&E and MT staining. Adipose tissue, identified by ORO staining, demonstrated both green and red AF (asterisk). PpIX fluorescence microscopic imaging confirmed cancer cell localization of PpIX (arrow), which was not observed during green AF imaging. Scale bar = 100 μm

Fig. 5

Ex vivo breast specimen fluorescence…

Fig. 5

Ex vivo breast specimen fluorescence in patients with and without 5-ALA. Representative white…

Fig. 5
Ex vivo breast specimen fluorescence in patients with and without 5-ALA. Representative white light and fluorescence images with corresponding biopsy-based H&E and fluorescence spectra from patients with invasive ductal carcinoma receiving A no 5-ALA, B 15 mg/kg 5-ALA, or C 30 mg/kg 5-ALA. (a) Biopsies were collected in areas inside (Bx1) the PA demarcated tumor (blue line) and outside the demarcated tumor (Bx2-4) on an adjacent slice of the specimen. Circular insets are digitally magnified images of the biopsy areas demonstrating the fluorescence color. (b) H&E-stained longitudinal biopsy sections were examined by a blinded pathologist (S.J.D.) for the presence of cancer. (c) Point spectroscopy was performed at the Bx1 location and smoothed fluorescence spectra in the region of PpIX emission (red box, 635 nm peak) are presented. D Representative chromaticity diagrams (CIE xyY displaying the average pixel color inside the demarcated tumor border and outside to normal tissue contrast from fluorescence images of specimens from patients described in parts AC of this figure. E Bar graph depicting the average vector distance between the average pixel color of the primary tumor and surrounding normal tissue. * p < 0.05, one-way ANOVA with multiple comparisons. Scale bar = 5 mm (a, white light and fluorescence images), 100 μm (a, inset), 500 μm (b, H&E sections). Bx, biopsy; IDC, invasive ductal carcinoma
Fig. 2
Fig. 2
Fluorescence imaging of 5-ALA-induced PpIX fluorescence in grossly obvious and grossly occult carcinoma. A Representative white light (top row) and fluorescence (bottom row) images of grossly obvious disease in sectioned lumpectomy specimens (a) and a clinically positive sentinel lymph node (c). The pathologist’s assistant (V.S., M.S., F.M.) demarcated tumor border (blue line) identified the grossly obvious tumor in the sectioned specimens. The surgeon (A.M.E., W.L.L.) identified the lymph node as grossly obvious for disease. B Representative white light (top row) and fluorescence (bottom row) images of grossly occult disease at the surface of an excised lumpectomy (a), in grossly sectioned specimens (b, c) and a sentinel lymph node (d) from patients with invasive ductal carcinoma with (a, d) or without (b, c) a DCIS component administered 15 mg/kg (b, c) or 30 mg/kg (a, d) 5-ALA HCl. Images represent tissue that was identified by the surgeon (A.M.E., W.L.L.) (a, d) or pathologist’s assistant (V.S., M.S., F.M.) (b, c) as grossly negative for the presence of cancer. (a) DCIS identified by fluorescence imaging at the lumpectomy margin. (b, c) Invasive carcinoma identified by fluorescence imaging on slices outside the grossly demarcated tumor. (d) Invasive carcinoma macro-metastases identified by fluorescence imaging in an excised sentinel lymph node. Scale bars = 5 mm
Fig. 3
Fig. 3
Detection of grossly occult sub-millimeter red fluorescence tumor foci. A WL and fluorescence images of a slice containing no clinically obvious disease from a patient who received 30 mg/kg 5-ALA HCl. Biopsies were collected in an area of focal red PpIX fluorescence (Bx1) and an adjacent area lacking PpIX fluorescence (Bx2). B H&E-stained longitudinal section of the Bx1 biopsy identified in A, which was determined to contain invasive ductal carcinoma by a blinded pathologist (S.J.D.). The imaged surface of the biopsy is indicated by the arrowheads. The area of tumor near the imaged surface measured 0.71 mm2. C H&E-stained longitudinal section of the Bx2 biopsy identified in A, which was determined to be negative for tumor by a blinded pathologist (S.J.D.). D White light and fluorescence images of slices of a lumpectomy from a patient who received 15 mg/kg 5-ALA HCl. A biopsy (Bx1) was collected from a small area of red fluorescence (inset digitally zoomed). E H&E-stained longitudinal section of the Bx1 identified in D, which was determined to contain DCIS > 2 mm below the imaged surface (arrowheads). Scale bar = 0.5 mm (A, D), 500 μm (B, C, E). WL, white light; FL, fluorescence; Bx, biopsy; IDC, invasive ductal carcinoma; DCIS, ductal carcinoma in situ
Fig. 4
Fig. 4
Cancer cell-specific localization of ALA-induced PpIX fluorescence. Representative A fluorescence microscopy and B corresponding histological images (bottom panel) from a biopsy collected inside the demarcated primary tumor boundary of a patient who received 30 mg/kg 5-ALA. The biopsy appeared red fluorescent with PRODIGI imaging. A Fluorescence microscopy was performed on cryosections cut from tumor core biopsies followed by B H&E, Masson’s Trichrome (MT), and Oil Red O (ORO) staining. Arrowheads depict green AF that was consistently observed in fibrous collagen tissue and in locations of necrosis, as confirmed with H&E and MT staining. Adipose tissue, identified by ORO staining, demonstrated both green and red AF (asterisk). PpIX fluorescence microscopic imaging confirmed cancer cell localization of PpIX (arrow), which was not observed during green AF imaging. Scale bar = 100 μm
Fig. 5
Fig. 5
Ex vivo breast specimen fluorescence in patients with and without 5-ALA. Representative white light and fluorescence images with corresponding biopsy-based H&E and fluorescence spectra from patients with invasive ductal carcinoma receiving A no 5-ALA, B 15 mg/kg 5-ALA, or C 30 mg/kg 5-ALA. (a) Biopsies were collected in areas inside (Bx1) the PA demarcated tumor (blue line) and outside the demarcated tumor (Bx2-4) on an adjacent slice of the specimen. Circular insets are digitally magnified images of the biopsy areas demonstrating the fluorescence color. (b) H&E-stained longitudinal biopsy sections were examined by a blinded pathologist (S.J.D.) for the presence of cancer. (c) Point spectroscopy was performed at the Bx1 location and smoothed fluorescence spectra in the region of PpIX emission (red box, 635 nm peak) are presented. D Representative chromaticity diagrams (CIE xyY displaying the average pixel color inside the demarcated tumor border and outside to normal tissue contrast from fluorescence images of specimens from patients described in parts AC of this figure. E Bar graph depicting the average vector distance between the average pixel color of the primary tumor and surrounding normal tissue. * p < 0.05, one-way ANOVA with multiple comparisons. Scale bar = 5 mm (a, white light and fluorescence images), 100 μm (a, inset), 500 μm (b, H&E sections). Bx, biopsy; IDC, invasive ductal carcinoma

References

    1. Chen K, Li S, Li Q, Zhu L, Liu Y, Song E, Su F. Breast-conserving surgery rates in breast cancer patients with different molecular subtypes: an observational study based on Surveillance, Epidemiology, and End Results (SEER) database. Medicine (Baltimore) 2016;95:e2593. doi: 10.1097/MD.0000000000002593.
    1. Porter G, Wagar B, Bryant H, Hewitt M, Wai E, Dabbs K, McFarlane A, Rahal R. Rates of breast cancer surgery in Canada from 2007/08 to 2009/10: retrospective cohort study. CMAJ open. 2014;2(2):E102–E108. doi: 10.9778/cmajo.20130025.
    1. Menes TS, Tartter PI, Bleiweiss I, Godbold JH, Estabrook A, Smith SR. The consequence of multiple re-excisions to obtain clear lumpectomy margins in breast cancer patients. Ann Surg Oncol. 2005;12(11):881–885. doi: 10.1245/ASO.2005.03.021.
    1. Hennigs A, Fuchs V, Sinn HP, Riedel F, Rauch G, Smetanay K, Golatta M, Domschke C, Schuetz F, Schneeweiss A, Sohn C, Heil J. Do patients after reexcision due to involved or close margins have the same risk of local recurrence as those after one-step breast-conserving surgery? Ann Surg Oncol. 2016;23(6):1831–1837. doi: 10.1245/s10434-015-5067-1.
    1. Ali AN, Vapiwala N, Guo M, Hwang WT, Harris EE, Solin LJ. The impact of re-excision and residual disease on local recurrence after breast conservation treatment for patients with early stage breast cancer. Clin Breast Cancer. 2011;11(6):400–405. doi: 10.1016/j.clbc.2011.08.003.
    1. Moran MS, Schnitt SJ, Giuliano AE, Harris JR, Khan SA, Horton J, Klimberg S, Chavez-MacGregor M, Freedman G, Houssami N, Johnson PL, Morrow M. Society of Surgical Oncology-American Society for Radiation Oncology consensus guideline on margins for breast-conserving surgery with whole-breast irradiation in stages I and II invasive breast cancer. Int J Radiat Oncol Biol Phys. 2014;88(3):553–564. doi: 10.1016/j.ijrobp.2013.11.012.
    1. Landercasper J, Attai D, Atisha D, Beitsch P, Bosserman L, Boughey J, Carter J, Edge S, Feldman S, Froman J, Greenberg C, Kaufman C, Morrow M, Pockaj B, Silverstein M, Solin L, Staley A, Vicini F, Wilke L, Yang W, Cody H., 3rd Toolbox to reduce lumpectomy reoperations and improve cosmetic outcome in breast cancer patients: The American Society of Breast Surgeons Consensus Conference. Ann Surg Oncol. 2015;22(10):3174–3183. doi: 10.1245/s10434-015-4759-x.
    1. Vandergrift JL, Niland JC, Theriault RL, Edge SB, Wong YN, Loftus LS, Breslin TM, Hudis CA, Javid SH, Rugo HS, Silver SM, Lepisto EM, Weeks JC. Time to adjuvant chemotherapy for breast cancer in National Comprehensive Cancer Network institutions. J Natl Cancer Inst. 2013;105(2):104–112. doi: 10.1093/jnci/djs506.
    1. Pataky RE, Baliski CR. Reoperation costs in attempted breast-conserving surgery: a decision analysis. Curr Oncol. 2016;23(5):314–321. doi: 10.3747/co.23.2989.
    1. Baliski CR, Pataky RE. Influence of the SSO/ASTRO margin reexcision guidelines on costs associated with breast-conserving surgery. Ann Surg Oncol. 2017;24(3):632–637. doi: 10.1245/s10434-016-5678-1.
    1. Orosco RK, Tapia VJ, Califano JA, Clary B, Cohen EEW, Kane C, Lippman SM, Messer K, Molinolo A, Murphy JD, Pang J, Sacco A, Tringale KR, Wallace A, Nguyen QT. Positive surgical margins in the 10 most common solid cancers. Sci Rep. 2018;8(1):5686. doi: 10.1038/s41598-018-23403-5.
    1. Zhan QH, Fu JQ, Fu FM, Zhang J, Wang C. Survival and time to initiation of adjuvant chemotherapy among breast cancer patients: a systematic review and meta-analysis. Oncotarget. 2018;9(2):2739–2751. doi: 10.18632/oncotarget.23086.
    1. Findlay-Shirras LJ, Outbih O, Muzyka CN, Galloway K, Hebbard PC, Nashed M. Predictors of residual disease after breast conservation surgery. Ann Surg Oncol. 2018;25(7):1936–1942. doi: 10.1245/s10434-018-6454-1.
    1. Fisher S, Yasui Y, Dabbs K, Winget M. Re-excision and survival following breast conserving surgery in early stage breast cancer patients: a population-based study. BMC Health Serv Res. 2018;18(1):94. doi: 10.1186/s12913-018-2882-7.
    1. Hughes L, Hamm J, McGahan C, Baliski C. Surgeon volume, patient age, and tumor-related factors influence the need for re-excision after breast-conserving surgery. Ann Surg Oncol. 2016;23(S5):656–664. doi: 10.1245/s10434-016-5602-8.
    1. Wilke LG, Czechura T, Wang C, Lapin B, Liederbach E, Winchester DP, Yao K. Repeat surgery after breast conservation for the treatment of stage 0 to II breast carcinoma: a report from the National Cancer Data Base, 2004-2010. JAMA Surg. 2014;149(12):1296–1305. doi: 10.1001/jamasurg.2014.926.
    1. Jeevan R, Cromwell DA, Trivella M, Lawrence G, Kearins O, Pereira J, Sheppard C, Caddy CM, van der Meulen JH. Reoperation rates after breast conserving surgery for breast cancer among women in England: retrospective study of hospital episode statistics. BMJ. 2012;345(jul12 2):e4505. doi: 10.1136/bmj.e4505.
    1. Langhans L, Jensen MB, Talman MM, Vejborg I, Kroman N, Tvedskov TF. Reoperation rates in ductal carcinoma in situ vs invasive breast cancer after wire-guided breast-conserving surgery. JAMA Surg. 2017;152(4):378–384. doi: 10.1001/jamasurg.2016.4751.
    1. Lovrics PJ, Gordon M, Cornacchi SD, Farrokhyar F, Ramsaroop A, Hodgson N, Quan ML, Wright F, Porter G. Practice patterns and perceptions of margin status for breast conserving surgery for breast carcinoma: National Survey of Canadian General Surgeons. Breast. 2012;21(6):730–734. doi: 10.1016/j.breast.2012.07.017.
    1. Parvez E, Hodgson N, Cornacchi SD, Ramsaroop A, Gordon M, Farrokhyar F, Porter G, Quan ML, Wright F, Lovrics PJ. Survey of American and Canadian general surgeons' perceptions of margin status and practice patterns for breast conserving surgery. Breast J. 2014;20(5):481–488. doi: 10.1111/tbj.12299.
    1. Thill M, Baumann K, Barinoff J. Intraoperative assessment of margins in breast conservative surgery-still in use? J Surg Oncol. 2014;110(1):15–20. doi: 10.1002/jso.23634.
    1. Biganzoli L, Marotti L, Hart CD, Cataliotti L, Cutuli B, Kuhn T, Mansel RE, Ponti A, Poortmans P, Regitnig P, van der Hage JA, Wengstrom Y, Rosselli Del Turco M. Quality indicators in breast cancer care: an update from the EUSOMA working group. Eur J Cancer. 2017;86:59–81. doi: 10.1016/j.ejca.2017.08.017.
    1. Butler-Henderson K, Lee AH, Price RI, Waring K. Intraoperative assessment of margins in breast conserving therapy: a systematic review. Breast. 2014;23(2):112–119. doi: 10.1016/j.breast.2014.01.002.
    1. Singh M, Singh G, Hogan KT, Atkins KA, Schroen AT. The effect of intraoperative specimen inking on lumpectomy re-excision rates. World J Surg Oncol. 2010;8(1):4. doi: 10.1186/1477-7819-8-4.
    1. Maloney BW, McClatchy DM, Pogue BW, Paulsen KD, Wells WA, Barth RJ. Review of methods for intraoperative margin detection for breast conserving surgery. J Biomed Opt. 2018;23(10):1–19. doi: 10.1117/1.JBO.23.10.100901.
    1. Gray RJ, Pockaj BA, Garvey E, Blair S. Intraoperative margin management in breast-conserving surgery: a systematic review of the literature. Ann Surg Oncol. 2018;25(1):18–27. doi: 10.1245/s10434-016-5756-4.
    1. Whitley MJ, Cardona DM, Lazarides AL, Spasojevic I, Ferrer JM, Cahill J, Lee CL, Snuderl M, Blazer DG, 3rd, Hwang ES, Greenup RA, Mosca PJ, Mito JK, Cuneo KC, Larrier NA, O'Reilly EK, Riedel RF, Eward WC, Strasfeld DB, Fukumura D, Jain RK, Lee WD, Griffith LG, Bawendi MG, Kirsch DG, Brigman BE. A mouse-human phase 1 co-clinical trial of a protease-activated fluorescent probe for imaging cancer. Sci Transl Med. 2016;8:320ra324. doi: 10.1126/scitranslmed.aad0293.
    1. Miampamba M, Liu J, Harootunian A, Gale AJ, Baird S, Chen SL, Nguyen QT, Tsien RY, Gonzalez JE. Sensitive in vivo visualization of breast cancer using ratiometric protease-activatable fluorescent imaging agent, AVB-620. Theranostics. 2017;7(13):3369–3386. doi: 10.7150/thno.20678.
    1. Zhang RR, Schroeder AB, Grudzinski JJ, Rosenthal EL, Warram JM, Pinchuk AN, Eliceiri KW, Kuo JS, Weichert JP. Beyond the margins: real-time detection of cancer using targeted fluorophores. Nat Rev Clin Oncol. 2017;14(6):347–364. doi: 10.1038/nrclinonc.2016.212.
    1. Nagaya T, Nakamura YA, Choyke PL, Kobayashi H. Fluorescence-guided surgery. Front Oncol. 2017;7:314. doi: 10.3389/fonc.2017.00314.
    1. DaCosta RS, Kulbatski I, Lindvere-Teene L, Starr D, Blackmore K, Silver JI, Opoku J, Wu YC, Medeiros PJ, Xu W, Xu L, Wilson BC, Rosen C, Linden R. Point-of-care autofluorescence imaging for real-time sampling and treatment guidance of bioburden in chronic wounds: first-in-human results. Plos one. 2015;10(3):e0116623. doi: 10.1371/journal.pone.0116623.
    1. Wu YC, Smith M, Chu A, Lindvere-Teene L, Starr D, Tapang K, Shekhman R, Wong O, Linden R, DaCosta RS. Handheld fluorescence imaging device detects subclinical wound infection in an asymptomatic patient with chronic diabetic foot ulcer: a case report. Int Wound J. 2015;13:449–453. doi: 10.1111/iwj.12451.
    1. Wu YC, Kulbatski I, Medeiros PJ, Maeda A, Bu J, Xu L, Chen Y, DaCosta RS. Autofluorescence imaging device for real-time detection and tracking of pathogenic bacteria in a mouse skin wound model: preclinical feasibility studies. J Biomed Opt. 2014;19(8):085002. doi: 10.1117/1.JBO.19.8.085002.
    1. Loh CS, MacRobert AJ, Bedwell J, Regula J, Krasner N, Bown SG. Oral versus intravenous administration of 5-aminolaevulinic acid for photodynamic therapy. Br J Cancer. 1993;68(1):41–51. doi: 10.1038/bjc.1993.284.
    1. Ohgari Y, Nakayasu Y, Kitajima S, Sawamoto M, Mori H, Shimokawa O, Matsui H, Taketani S. Mechanisms involved in delta-aminolevulinic acid (ALA)-induced photosensitivity of tumor cells: relation of ferrochelatase and uptake of ALA to the accumulation of protoporphyrin. Biochem Pharmacol. 2005;71(1-2):42–49. doi: 10.1016/j.bcp.2005.10.019.
    1. Kobayashi H, Choyke PL. Target-cancer-cell-specific activatable fluorescence imaging probes: rational design and in vivo applications. Acc Chem Res. 2011;44(2):83–90. doi: 10.1021/ar1000633.
    1. Krieg RC, Messmann H, Rauch J, Seeger S, Knuechel R. Metabolic characterization of tumor cell-specific protoporphyrin IX accumulation after exposure to 5-aminolevulinic acid in human colonic cells. Photochem Photobiol. 2002;76(5):518–525. doi: 10.1562/0031-8655(2002)076<0518:MCOTCS>;2.
    1. Valdes PA, Leblond F, Kim A, Harris BT, Wilson BC, Fan X, Tosteson TD, Hartov A, Ji S, Erkmen K, Simmons NE, Paulsen KD, Roberts DW. Quantitative fluorescence in intracranial tumor: implications for ALA-induced PpIX as an intraoperative biomarker. J Neurosurg. 2011;115(1):11–17. doi: 10.3171/2011.2.JNS101451.
    1. Valdes PA, Kim A, Brantsch M, Niu C, Moses ZB, Tosteson TD, Wilson BC, Paulsen KD, Roberts DW, Harris BT. delta-aminolevulinic acid-induced protoporphyrin IX concentration correlates with histopathologic markers of malignancy in human gliomas: the need for quantitative fluorescence-guided resection to identify regions of increasing malignancy. Neuro Oncol. 2011;13:846–856. doi: 10.1093/neuonc/nor086.
    1. Valdes PA, Bekelis K, Harris BT, Wilson BC, Leblond F, Kim A, Simmons NE, Erkmen K, Paulsen KD, Roberts DW. 5-Aminolevulinic acid-induced protoporphyrin IX fluorescence in meningioma: qualitative and quantitative measurements in vivo. Neurosurgery. 2014;10(Suppl 1):74–82.
    1. Millon SR, Ostrander JH, Yazdanfar S, Brown JQ, Bender JE, Rajeha A, Ramanujam N. Preferential accumulation of 5-aminolevulinic acid-induced protoporphyrin IX in breast cancer: a comprehensive study on six breast cell lines with varying phenotypes. J Biomed Opt. 2010;15(1):018002. doi: 10.1117/1.3302811.
    1. Palasuberniam P, Yang X, Kraus D, Jones P, Myers KA, Chen B. ABCG2 transporter inhibitor restores the sensitivity of triple negative breast cancer cells to aminolevulinic acid-mediated photodynamic therapy. Sci Rep. 2015;18:13298. doi: 10.1038/srep13298.
    1. Lakomkin N, Hadjipanayis CG. Fluorescence-guided surgery for high-grade gliomas. J Surg Oncol. 2018;118(2):356–361. doi: 10.1002/jso.25154.
    1. Nakai Y, Inoue K, Tsuzuki T, Shimamoto T, Shuin T, Nagao K, Matsuyama H, Oyama M, Furuse H, Ozono S, Miyake M, Fujimoto K. Oral 5-aminolevulinic acid-mediated photodynamic diagnosis using fluorescence cystoscopy for non-muscle-invasive bladder cancer: A multicenter phase III study. Int J Urol. 2018;25(8):723–729. doi: 10.1111/iju.13718.
    1. Inoue K, Anai S, Fujimoto K, Hirao Y, Furuse H, Kai F, Ozono S, Hara T, Matsuyama H, Oyama M, Ueno M, Fukuhara H, Narukawa M, Shuin T. Oral 5-aminolevulinic acid mediated photodynamic diagnosis using fluorescence cystoscopy for non-muscle-invasive bladder cancer: a randomized, double-blind, multicentre phase II/III study. Photodiagnosis Photodyn Ther. 2015;12(2):193–200. doi: 10.1016/j.pdpdt.2015.03.008.
    1. Tsuruki ES, Saito Y, Abe S, Takamaru H, Yamada M, Sakamoto T, Nakajima T, Matsuda T, Sekine S, Taniguchi H. Evaluating the efficacy and safety of a novel endoscopic fluorescence imaging modality using oral 5-aminolevulinic acid for colorectal tumors. Endosc Int Open. 2016;4(1):E30–E35. doi: 10.1055/s-0041-110432.
    1. Kitada M, Ohsaki Y, Yasuda S, Abe M, Takahashi N, Okazaki S, Ishibashi K, Hayashi S. Photodynamic diagnosis of visceral pleural invasion of lung cancer with a combination of 5-aminolevulinic acid and autofluorescence observation systems. Photodiagnosis Photodyn Ther. 2017;20:10–15. doi: 10.1016/j.pdpdt.2017.08.013.
    1. Ladner DP, Steiner RA, Allemann J, Haller U, Walt H. Photodynamic diagnosis of breast tumours after oral application of aminolevulinic acid. Br J Cancer. 2001;84(1):33–37. doi: 10.1054/bjoc.2000.1532.
    1. Matoba Y, Banno K, Kisu I, Aoki D. Clinical application of photodynamic diagnosis and photodynamic therapy for gynecologic malignant diseases: a review. Photodiagnosis Photodyn Ther. 2018;24:52–7. 10.1016/j.pdpdt.2018.08.014.
    1. Messmann H. 5-Aminolevulinic acid-induced protoporphyrin IX for the detection of gastrointestinal dysplasia. Gastrointest Endosc Clin N Am. 2000;10(3):497–512. doi: 10.1016/S1052-5157(18)30119-3.
    1. Leunig A, Betz CS, Mehlmann M, Stepp H, Arbogast S, Grevers G, Baumgartner R. Detection of squamous cell carcinoma of the oral cavity by imaging 5-aminolevulinic acid-induced protoporphyrin IX fluorescence. Laryngoscope. 2000;110(1):78–83. doi: 10.1097/00005537-200001000-00015.
    1. Wang I, Clemente LP, Pratas RM, Cardoso E, Clemente MP, Montan S, Svanberg S, Svanberg K. Fluorescence diagnostics and kinetic studies in the head and neck region utilizing low-dose delta-aminolevulinic acid sensitization. Cancer Lett. 1999;135(1):11–19. doi: 10.1016/s0304-3835(98)00271-7.
    1. Vanseviciute R, Venius J, Zukovskaja O, Kanopiene D, Letautiene S, Rotomskis R. 5-aminolevulinic-acid-based fluorescence spectroscopy and conventional colposcopy for in vivo detection of cervical pre-malignancy. BMC Womens Health. 2015;15(1):35. doi: 10.1186/s12905-015-0191-4.
    1. Malik E, Berg C, Meyhofer-Malik A, Buchweitz O, Moubayed P, Diedrich K. Fluorescence diagnosis of endometriosis using 5-aminolevulinic acid. Surg Endosc. 2000;14(5):452–455. doi: 10.1007/s004640000160.
    1. Kitada M, Ohsaki Y, Matsuda Y, Hayashi S, Ishibashi K. Photodynamic diagnosis of pleural malignant lesions with a combination of 5-aminolevulinic acid and intrinsic fluorescence observation systems. BMC Cancer. 2015;15(1):174. doi: 10.1186/s12885-015-1194-0.
    1. Kanick SC, Davis SC, Zhao Y, Hasan T, Maytin EV, Pogue BW, Chapman MS. Dual-channel red/blue fluorescence dosimetry with broadband reflectance spectroscopic correction measures protoporphyrin IX production during photodynamic therapy of actinic keratosis. J Biomed Opt. 2014;19(7):75002. doi: 10.1117/1.JBO.19.7.075002.
    1. Ozog DM, Rkein AM, Fabi SG, Gold MH, Goldman MP, Lowe NJ, Martin GM, Munavalli GS. Photodynamic therapy: a clinical consensus guide. Dermatol Surg. 2016;42(7):804–827. doi: 10.1097/DSS.0000000000000800.
    1. A. Nabavi, H. Thurm, B. Zountsas, T. Pietsch, H. Lanfermann, U. Pichlmeier, M. Mehdorn, A. L. A. R. G. S. Group, Five-aminolevulinic acid for fluorescence-guided resection of recurrent malignant gliomas: a phase ii study. Neurosurgery. 2009;65:1070-1076. discussion 1076-1077
    1. Stummer W, Pichlmeier U, Meinel T, Wiestler OD, Zanella F, Reulen HJ. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 2006;7(5):392–401. doi: 10.1016/S1470-2045(06)70665-9.
    1. Stummer W, Stepp H, Wiestler OD, Pichlmeier U. Randomized, prospective double-blinded study comparing 3 different doses of 5-aminolevulinic acid for fluorescence-guided resections of malignant gliomas. Neurosurgery. 2017;81(2):230–239. doi: 10.1093/neuros/nyx074.
    1. Stummer W, Tonn JC, Mehdorn HM, Nestler U, Franz K, Goetz C, Bink A, Pichlmeier U. Counterbalancing risks and gains from extended resections in malignant glioma surgery: a supplemental analysis from the randomized 5-aminolevulinic acid glioma resection study. Clinical article. J Neurosurg. 2011;114(3):613–623. doi: 10.3171/2010.3.JNS097.
    1. Development Corp NX. Gleolan (aminolevulinic acid hydrochloride): Highlights of prescribing information. 2019.
    1. Webber J, Kessel D, Fromm D. Plasma levels of protoporphyrin IX in humans after oral administration of 5-aminolevulinic acid. J Photochem Photobiol B. 1997;37(1-2):151–153. doi: 10.1016/S1011-1344(96)07348-4.
    1. Cozzens JW, Lokaitis BC, Moore BE, Amin DV, Espinosa JA, MacGregor M, Michael AP, Jones BA. A Phase 1 dose-escalation study of oral 5-aminolevulinic acid in adult patients undergoing resection of a newly diagnosed or recurrent high-grade glioma. Neurosurgery. 2017;81(1):46–55. doi: 10.1093/neuros/nyw182.
    1. Webber J, Kessel D, Fromm D. Side effects and photosensitization of human tissues after aminolevulinic acid. J Surg Res. 1997;68(1):31–37. doi: 10.1006/jsre.1997.5004.
    1. Olivotto I, Levine M. Clinical practice guidelines for the care and treatment of breast cancer: the management of ductal carcinoma in situ (summary of the 2001 update) CMAJ. 2001;165(7):912–913.
    1. Hagiya AS, Etman A, Siddiqi IN, Cen S, Matcuk GR, Jr, Brynes RK, Salama ME. Digital image analysis agrees with visual estimates of adult bone marrow trephine biopsy cellularity. Int J Lab Hematol. 2018;40(2):209–214. doi: 10.1111/ijlh.12768.
    1. Monici M. Cell and tissue autofluorescence research and diagnostic applications. Biotechnol Annu Rev. 2005;11:227–256. doi: 10.1016/S1387-2656(05)11007-2.
    1. Priye A, Ball CS, Meagher RJ. Colorimetric-luminance readout for quantitative analysis of fluorescence signals with a smartphone CMOS sensor. Anal Chem. 2018;90(21):12385–12389. doi: 10.1021/acs.analchem.8b03521.
    1. Morrow M, Van Zee KJ, Solin LJ, Houssami N, Chavez-MacGregor M, Harris JR, Horton J, Hwang S, Johnson PL, Marinovich ML, Schnitt SJ, Wapnir I, Moran MS. Society of Surgical Oncology-American Society for Radiation Oncology-American Society of Clinical Oncology Consensus Guideline on margins for breast-conserving surgery with whole-breast irradiation in ductal carcinoma in situ. Ann Surg Oncol. 2016;23(12):3801–3810. doi: 10.1245/s10434-016-5449-z.
    1. M. Morrow, P. Abrahamse, T. P. Hofer, K. C. Ward, A. S. Hamilton, A. W. Kurian, S. J. Katz, R. Jagsi, Trends in reoperation after initial lumpectomy for breast cancer: addressing overtreatment in surgical management. JAMA Oncol. 2017;3(10):1352-7. 10.1001/jamaoncol.2017.0774.
    1. DeSnyder SM, Hunt KK, Smith BD, Moran MS, Klimberg S, Lucci A. Assessment of practice patterns following publication of the SSO-ASTRO consensus guideline on margins for breast-conserving therapy in stage I and II invasive breast cancer. Ann Surg Oncol. 2015;22(10):3250–3256. doi: 10.1245/s10434-015-4666-1.
    1. K. J. L. Blair, M., Re-excision rates following breast conserving therapy: a single institutions experience over ten years. Marshal J Med. 2017;3:68-74
    1. Merrill AL, Coopey SB, Tang R, McEvoy MP, Specht MC, Hughes KS, Gadd MA, Smith BL. Implications of new lumpectomy margin guidelines for breast-conserving surgery: changes in reexcision rates and predicted rates of residual tumor. Ann Surg Oncol. 2016;23(3):729–734. doi: 10.1245/s10434-015-4916-2.
    1. Chung A, Gangi A, Amersi F, Bose S, Zhang X, Giuliano A. Impact of Consensus Guidelines by the Society of Surgical Oncology and the American Society for Radiation Oncology on Margins for Breast-Conserving Surgery in Stages 1 and 2 Invasive Breast Cancer. Ann Surg Oncol. 2015;22(Suppl 3):S422–S427. doi: 10.1245/s10434-015-4829-0.
    1. Rosenberger LH, Mamtani A, Fuzesi S, Stempel M, Eaton A, Morrow M, Gemignani ML. Early adoption of the SSO-ASTRO Consensus Guidelines on margins for breast-conserving surgery with whole-breast irradiation in stage i and ii invasive breast cancer: initial experience from Memorial Sloan Kettering Cancer Center. Ann Surg Oncol. 2016;23(10):3239–3246. doi: 10.1245/s10434-016-5397-7.
    1. Schulman AM, Mirrielees JA, Leverson G, Landercasper J, Greenberg C, Wilke LG. Reexcision surgery for breast cancer: an analysis of the American Society of Breast Surgeons (ASBrS) MasterySM database following the SSO-ASTRO “No Ink on Tumor” Guidelines. Ann Surg Oncol. 2017;24(1):52–58. doi: 10.1245/s10434-016-5516-5.
    1. Heelan Gladden AA, Sams S, Gleisner A, Finlayson C, Kounalakis N, Hosokawa P, Brown R, Chong T, Mathes D, Murphy C. Re-excision rates after breast conserving surgery following the 2014 SSO-ASTRO guidelines. Am J Surg. 2017;214(6):1104–1109. doi: 10.1016/j.amjsurg.2017.08.023.
    1. Tang SS, Kaptanis S, Haddow JB, Mondani G, Elsberger B, Tasoulis MK, Obondo C, Johns N, Ismail W, Syed A, Kissias P, Venn M, Sundaramoorthy S, Irwin G, Sami AS, Elfadl D, Baggaley A, Remoundos DD, Langlands F, Charalampoudis P, Barber Z, Hamilton-Burke WLS, Khan A, Sirianni C, Merker LAG, Saha S, Lane RA, Chopra S, Dupre S, Manning AT, John ERS, Musbahi A, Dlamini N, McArdle CL, Wright C, Murphy JO, Aggarwal R, Dordea M, Bosch K, Egbeare D, Osman H, Tayeh S, Razi F, Iqbal J, Ledwidge SFC, Albert V. Current margin practice and effect on re-excision rates following the publication of the SSO-ASTRO consensus and ABS consensus guidelines: a national prospective study of 2858 women undergoing breast-conserving therapy in the UK and Ireland. Eur J Cancer. 2017;84:315–324. doi: 10.1016/j.ejca.2017.07.032.
    1. Chagpar AB, Horowitz NR, Killelea BK, Tsangaris T, Longley P, Grizzle S, Loftus M, Li F, Butler M, Stavris K, Yao X, Harigopal M, Bossuyt V, Lannin DR, Pusztai L, Davidoff AJ, Gross CP. Economic impact of routine cavity margins versus standard partial mastectomy in breast cancer patients: results of a randomized controlled trial. Ann Surg. 2017;265(1):39–44. doi: 10.1097/SLA.0000000000001799.
    1. Chagpar AB, Killelea BK, Tsangaris TN, Butler M, Stavris K, Li F, Yao X, Bossuyt V, Harigopal M, Lannin DR, Pusztai L, Horowitz NR. A randomized, controlled trial of cavity shave margins in breast cancer. N Engl J Med. 2015;373(6):503–510. doi: 10.1056/NEJMoa1504473.
    1. Molinaro AM. Diagnostic tests: how to estimate the positive predictive value. Neurooncol Pract. 2015;2(4):162–166. doi: 10.1093/nop/npv030.
    1. Lalkhen AG, McCluskey A. Clinical tests: sensitivity and specificity. BJA Educattion. 2008;8:221–223.
    1. Coble J, Reid V. Achieving clear margins. Directed shaving using MarginProbe, as compared to a full cavity shave approach. Am J Surg. 2017;213:627–630. doi: 10.1016/j.amjsurg.2016.12.019.
    1. Pappo I, Spector R, Schindel A, Morgenstern S, Sandbank J, Leider LT, Schneebaum S, Lelcuk S, Karni T. Diagnostic performance of a novel device for real-time margin assessment in lumpectomy specimens. J Surg Res. 2010;160(2):277–281. doi: 10.1016/j.jss.2009.02.025.
    1. Schnabel F, Boolbol SK, Gittleman M, Karni T, Tafra L, Feldman S, Police A, Friedman NB, Karlan S, Holmes D, Willey SC, Carmon M, Fernandez K, Akbari S, Harness J, Guerra L, Frazier T, Lane K, Simmons RM, Estabrook A, Allweis T. A randomized prospective study of lumpectomy margin assessment with use of MarginProbe in patients with nonpalpable breast malignancies. Ann Surg Oncol. 2014;21(5):1589–1595. doi: 10.1245/s10434-014-3602-0.
    1. Omoto K, Matsuda R, Nakagawa I, Motoyama Y, Nakase H. False-positive inflammatory change mimicking glioblastoma multiforme under 5-aminolevulinic acid-guided surgery: A case report. Surg Neurol Int. 2018;9:49. doi: 10.4103/sni.sni_473_17.
    1. Utsuki S, Oka H, Sato S, Shimizu S, Suzuki S, Tanizaki Y, Kondo K, Miyajima Y, Fujii K. Histological examination of false positive tissue resection using 5-aminolevulinic acid-induced fluorescence guidance. Neurol Med Chir (Tokyo) 2007;47:210–213. doi: 10.2176/nmc.47.210.
    1. Wilbers E, Hargus G, Wolfer J, Stummer W. Usefulness of 5-ALA (Gliolan(R))-derived PPX fluorescence for demonstrating the extent of infiltration in atypical meningiomas. Acta Neurochir (Wien) 2014;156(10):1853–1854. doi: 10.1007/s00701-014-2148-z.
    1. Nakhlis F. How do we approach benign proliferative lesions? Curr Oncol Rep. 2018;20(4):34. doi: 10.1007/s11912-018-0682-1.
    1. DaCosta RS, Andersson H, Wilson BC. Molecular fluorescence excitation-emission matrices relevant to tissue spectroscopy. Photochem Photobiol. 2003;78(4):384–392. doi: 10.1562/0031-8655(2003)0780384:;2.
    1. Vera-Badillo FE, Napoleone M, Ocana A, Templeton AJ, Seruga B, Al-Mubarak M, AlHashem H, Tannock IF, Amir E. Effect of multifocality and multicentricity on outcome in early stage breast cancer: a systematic review and meta-analysis. Breast Cancer Res Treat. 2014;146(2):235–244. doi: 10.1007/s10549-014-3018-3.
    1. Cox TR, Erler JT. Molecular pathways: connecting fibrosis and solid tumor metastasis. Clin Cancer Res. 2014;20(14):3637–3643. doi: 10.1158/1078-0432.CCR-13-1059.
    1. Macmillan RD, McCulley SJ. Oncoplastic breast surgery: what, when and for whom? Curr Breast Cancer Rep. 2016;8(2):112–117. doi: 10.1007/s12609-016-0212-9.
    1. Dickson-Witmer D, Bleznak AD, Kennedy JS, Stewart AK, Palis BE, Bailey L, Laidley AL, Penman EJ. Breast cancer care in the community: challenges, opportunities, and outcomes. Surg Oncol Clin N Am. 2011;20:555–580, ix. doi: 10.1016/j.soc.2011.01.007.
    1. Rick K, Sroka R, Stepp H, Kriegmair M, Huber RM, Jacob K, Baumgartner R. Pharmacokinetics of 5-aminolevulinic acid-induced protoporphyrin IX in skin and blood. J Photochem Photobiol B. 1997;40(3):313–319. doi: 10.1016/S1011-1344(97)00076-6.
    1. Korb ML, Hartman YE, Kovar J, Zinn KR, Bland KI, Rosenthal EL. Use of monoclonal antibody-IRDye800CW bioconjugates in the resection of breast cancer. J Surg Res. 2014;188(1):119–128. doi: 10.1016/j.jss.2013.11.1089.
    1. Lamberts LE, Koch M, de Jong JS, Adams A, Glatz J, Tranendonk MEG, Terwisscha van Scheltinga AGT, Jansen L, de Vries J, Lub-de Hooge MN, Schroder CP, Jorritsma-Smit A, Linssen MD, de Boer E, van der Vegt B, Nagengast WB, Elias SG, Oliveira S, Witkamp A, Mali WPTM, van der Wall E, Van Diest PJ, de Vries EG, Ntziachristos V, van Dam GM. Tumor-specific uptake of fluorescent bevacizumab-IRDye800CW microdosing in patients with primary breast cancer: a phase I feasibility study. Clin Cancer Res. 2016;23(11):2730–41. 10.1158/1078-0432.CCR-16-0437.
    1. Tummers WS, Warram JM, Tipirneni KE, Fengler J, Jacobs P, Shankar L, Henderson L, Ballard B, Pfefer TJ, Pogue BW, Weichert JP, Bouvet M, Sorger J, Contag CH, Frangioni JV, Tweedle MF, Basilion JP, Gambhir SS, Rosenthal EL. Regulatory aspects of optical methods and exogenous targets for cancer detection. Cancer Res. 2017;77(9):2197–2206. doi: 10.1158/0008-5472.CAN-16-3217.

Source: PubMed

3
Abonnieren