Skip to main content
Erschienen in: Annals of Surgical Oncology 1/2023

Open Access 04.09.2022 | Translational Research

Proof of Principle of Combining Fluorescence-Guided Surgery with Photoimmunotherapy to Improve the Outcome of Pancreatic Cancer Therapy in an Orthotopic Mouse Model

verfasst von: Hiroto Nishino, MD, PhD, Michael A. Turner, MD, Siamak Amirfakhri, PhD, DVM, Thinzar M. Lwin, MD, MS, Mojgan Hosseini, MD, Bernhard B. Singer, PhD, Robert M. Hoffman, PhD, Michael Bouvet, MD

Erschienen in: Annals of Surgical Oncology | Ausgabe 1/2023

Abstract

Background

Pancreatic cancer is a recalcitrant disease in which R0 resection is often not achieved owing to difficulty in visualization of the tumor margins and proximity of adjacent vessels. To improve outcomes, we have developed fluorescence-guided surgery (FGS) and photoimmunotherapy (PIT) using a fluorescent tumor-specific antibody.

Methods

Nude mice received surgical orthotopic implantation (SOI) of the human pancreatic cancer cell line BxPC-3 expressing green fluorescent protein. An anti-carcinoembryonic antigen-related cell adhesion molecule (CEACAM) monoclonal antibody (6G5j) was conjugated to the 700-nm fluorescent dye IR700DyeDX (6G5j-IR700DX). Three weeks after SOI, 16 mice received 50 μg 6G5j-IR700DX via the tail vein 24 h before surgery and were randomized to two groups: FGS-only (n = 8) and FGS + PIT (n = 8). All tumors were imaged with the Pearl Trilogy imaging system and resected under the guidance of the FLARE imaging system. The FGS + PIT group received PIT of the post-surgical bed at an intensity of 150 mW/cm2 for 30 min. Mice were sacrificed 4 weeks after initial surgery, and tumors were imaged with a Dino-Lite digital microscope, excised, and weighed.

Results

The 6G5j-IR700DX dye illuminated the orthotopic pancreatic tumors for FGS and PIT. The metastatic recurrence rate was 100.0% for FGS-only and 25.0% for FGS + PIT (p = 0.007). The average total recurrent tumor weight was 2370.3 ± 1907.8 mg for FGS-only and 705.5 ± 1200.0 mg for FGS + PIT (p = 0.039).

Conclusions

FGS and adjuvant PIT can be combined by using a single antibody–fluorophore conjugate to significantly reduce the frequency of pancreatic cancer recurrence.
Hinweise
This work was presented at the Society of Surgical Oncology (SSO) 2022 International Conference on Surgical Cancer Care, Dallas, TX, 9–12 March 2022.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Pancreatic cancer is a recalcitrant disease and one of the most fatal cancers with a 5-year survival rate of only about 9%.1,2 Even after potentially curative surgery, 80% of patients will suffer recurrence and die of the disease.3 This is due, in part, to the difficulty of visualizing surgical margins, the difficulty in performing extended resection owing to the proximity of adjacent blood vessels, especially in the uncinate process, and the presence of microscopic metastatic disease.
Fluorescence-guided surgery (FGS), which can use tumor-specific antibodies conjugated with fluorescent dyes to visualize primary tumors and metastases, is making progress in preclinical and clinical trials.4,5 In our laboratory, we have demonstrated the usefulness of FGS with an anti-carcinoembryonic antigen (CEA) antibody conjugated to a 650-nm fluorophore in a pancreatic cancer patient-derived orthotopic xenograft (PDOX) model.6 We have also shown the efficacy of a humanized anti-CEA antibody conjugated to an 800-nm fluorescent dye that labels pancreatic cancer in a PDOX mouse model.7
Photoimmunotherapy (PIT) using a target-specific photosensitizer based on a near-infrared (NIR) phthalocyanine dye conjugated to monoclonal antibodies has been developed for cancer therapy.8 PIT decreases the number of cancer cells and enhances host immune responses against the tumor. PIT has been used to treat local tumors as well as metastasis and prevents recurrence.9 In our laboratory, we have reported the usefulness of PIT with an anti-CEA antibody conjugated to 700-nm dye for pancreatic cancer nude mouse models.1012 Initially, only PIT was used for the treatment of pancreatic cancer10 or bright-light surgery along with PIT.11
Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) are members of the carcinoembryonic antigen (CEA) gene family and the immunoglobulin superfamily.13 We have reported on the labeling of liver metastases from colorectal cancer using an anti-CEACAM antibody conjugated to a 700-nm fluorescent dye in a colon cancer liver metastases PDOX mouse model.14
The aim of the present study was to perform FGS and adjuvant PIT using a single antibody–fluorophore conjugate and to evaluate whether PIT can improve the prognosis of pancreatic cancer after FGS in an orthotopic mouse model.

Methods

Nude Mice

Nude (nu/nu) mice, aged 4–6 weeks, were purchased from Jackson Lab (Bar Harbor, ME). The animals were fed an autoclaved laboratory diet. All surgical procedures were performed under anesthesia by intramuscular injection of ketamine, xylazine, and acepromazine reconstituted in phosphate-buffered saline (PBS). Mice were treated with buprenorphine for pain control after surgical procedures. At the conclusion of the study, mice were euthanized with CO2 inhalation, which was confirmed with cervical dislocation. All studies were approved by the San Diego Veterans Administration Medical Center Institutional Animal Care and Use Committee (IACUC, animal use protocol A17-020).

Cell Culture

Human pancreatic cancer cell line BxPC-3 expressing green fluorescent protein (GFP) (AntiCancer Inc., San Diego, CA) was maintained in Roswell Park Memorial Institute (RPMI) 1640 medium (GIBCO-BRL, Grand Island, NY). The medium was supplemented with 10% fetal calf serum (Hyclone, Logan, UT), 1% l-glutamine, and 1% penicillin/streptomycin (GIBCO-BRL). The cells were incubated at 37 °C in a 5% CO2 incubator.

Cell Line Tumor

BxPC-3-GFP cells (1 × 106) were initially injected subcutaneously in bilateral flanks of nude mice. Resulting tumors were harvested and sectioned into small pieces for surgical orthotopic implantation (SOI) to the pancreas.

SOI of Pancreas Cancer to the Pancreas

The procedure of pancreatic cancer SOI was previously described.15,16 Briefly, a 10-mm transverse incision was made on the left flank of the mouse through the skin and peritoneum. The tail of the pancreas and spleen was exposed, and a single 1-mm3 tumor fragment from subcutaneous tumors was sutured to the tail of the pancreas using 8–0 surgical sutures (Ethicon Inc., Sommerville, NJ). The tail of the pancreas and spleen was returned to the abdomen, and the incision was closed with 6–0 surgical sutures (Ethicon Inc., Sommerville, NJ).

Administration of Fluorescent Agents

Anti-CEACAM monoclonal antibody (6G5j) was a kind gift from Dr. B. B. Singer, Institute for Anatomy, Essen, Germany. The antibody was conjugated to a near-infrared dye IR700DyeDX NHS ester (LI-COR Biosciences Inc., Lincoln, NE) to establish 6G5j-IR700DX as per previously described methods.14,17

Western Blotting

Tumor lysates were made using normal human pancreas tissue and the pancreatic cancer cell line BxPC-3-GFP xenograft. Western blotting was performed as previously described.18 Protein samples were isolated and transferred to Trans-Blot Turbo mini nitrocellulose membranes (Bio-Rad, Hercules, CA, Cat. 1704270) using the Bio-Rad Trans-Blot Turbo transfer system. The membrane was then incubated with the IRDye700DX-conjugated antibody at 4 °C overnight. The membrane was scanned with an Odyssey infrared imaging system (model 9120; LI-COR), and detection and quantification of band intensities were conducted using Image Studio Lite software (version 5.2; LI-COR). β-Actin was used as standard.

Time Course of Fluorescence Intensity in Orthotopic Tumors

Three weeks after SOI, mice were administered 50 μg 6G5j-IR700DX via tail vein injection. In vivo imaging at 700-nm wavelength was performed daily up to 72 h using the Pearl Trilogy small animal imaging system (LI-COR). Image analysis was performed using Image Studio software small animal imaging analysis (LI-COR). The pancreas was set as the background, and an area of interest around the tumor fluorescence was automatically provided by the system, with a minimum of 250 pixels and at least 2.5 standard deviations from the background signal. The tumor-to-pancreas ratio (TPR) was calculated for each mouse by dividing the maximal fluorescence intensity (MFI) of the pancreatic tumor by the MFI of the adjacent pancreas.

Intravital Static Imaging

Mice were anesthetized prior to imaging, and a laparotomy was performed to expose the pancreas. For 700-nm intravital static imaging, the Pearl Trilogy small animal fluorescence imaging system was used.

FGS and PIT under Guidance of Intravital Dynamic Imaging

After confirmation of tumor engraftment, mice were randomized to two groups: FGS-only and FGS + PIT. Each treatment arm included eight tumor-bearing mice. A schematic diagram of the experimental protocol is shown in Fig. 1. Mice received 50 μg 6G5j-IR700DX via tail vein 24 h before surgery. For surgery, a 10-mm transverse incision was made on the left flank of the mouse through the skin and peritoneum and the tail of the pancreas and spleen was exposed through this incision. For intravital dynamic imaging, an overlay mode (bright light, 700 nm) with the FLARE imaging system (Curadel, MA) was used. A Dino-Lite digital microscope (AnMo Electronics Corporation, Taiwan) was also used to visualize GFP expression. FGS was performed after intravital static imaging. Manual pressure was applied to the resection site for hemostasis at the conclusion of the procedure.
In the FGS + PIT group, photoimmunotherapy was performed as described previously.12,19 The resection bed was then irradiated with light from a red light-emitting diode at 690-nm wavelength (NIR laser; Ultralasers, Inc., Newmarket, Ontario, Canada) and power density of 150 mW/cm2, as measured by an optical power meter (PM 100; Thorlabs, Inc., Newton, NJ) for 30 min. The distance from the laser source to the tumor was standardized at 15 cm for each mouse. Surrounding normal tissues were protected by aluminum foil during PIT.
After treatment completion, the remnant pancreas was gently returned to the peritoneal cavity, the abdominal wall and skin were closed with 6–0 surgical sutures (Ethicon Inc., Sommerville, NJ), and the mice were allowed to recover in their cages. Resected tumors were then sectioned from paraffin blocks, and hematoxylin and eosin (H&E) staining was performed at the University of California San Diego Moore’s Cancer Center Histology Core. Paraffin sections were imaged on a Lionheart FX microscope (Agilent Technologies Inc., Santa Clara, CA) to obtain bright-light images of H&E staining.

Comparison of FGS-Only and FGS + PIT

To assess for postoperative recurrence, mice were sacrificed 4 weeks after initial surgery and underwent necropsy. Recurrent tumors expressing GFP were imaged with a Dino-Lite digital microscope, excised, and weighed in both groups. Local recurrence was defined as being located at the tail of the pancreas, where the tumor was resected, and having continuity with the pancreas, while other recurrences were defined as metastatic recurrence.

Statistical Analysis

Statistical analysis was performed using SAS software (JMP 14.2.0; SAS Institute Inc., Cary, NC). Comparisons between categorical variables were analyzed with Fisher’s exact test. Continuous variables are expressed as mean ± standard deviation, and comparisons between continuous variables were determined with the Mann–Whitney U test. A p-value < 0.05 was considered statistically significant for all comparisons.

Results

Determination of CEACAM Expression

Western blotting was performed for normal pancreas and pancreatic cancer cell line BxPC-3-GFP, demonstrating the expression of CEACAM protein (Fig. 2). Normal pancreas did not express CEACAM protein.

Tumor Labeling Time Course with 6G5j-IR700DX

Intravital imaging of BxPC-3-GFP pancreatic cancer xenografts showed that tumors were visualized using 6G5j-IR700DX (Fig. 3A). Peak MFI and TPR was observed at 24 h (Fig. 3B).

Labeling of Pancreatic Cancer for FGS and PIT

The 6G5j-IR700DX brightly illuminated the orthotopic pancreatic tumors for FGS and PIT visualized with the Pearl Trilogy small animal imaging system and the FLARE imaging system. GFP-expression of the pancreatic cancer could also be detected with the Dino-Lite digital microscope (Fig. 4).

FGS of the Pancreatic Cancer

The resection line was determined by the tumor margin on the basis of the fluorescence signal at 700 nm from 6G5j-IR700DX. The pancreatic tumor was resected with scissors, guided by fluorescence at all times (Fig. 5A–C). Grossly negative margins were obtained, and microscopic residual cancer was not visualized (Fig. 4). Representative bright-light and fluorescent images of the excised tumor are shown in Fig. 5D. The harvested pancreatic cancer was shown in H&E-labeled sections to have invaded the normal pancreas (Fig. 5E).

Comparison of FGS-Only and FGS + PIT

A representative image of recurrent tumors in the FGS-only group at 4 weeks after surgery is shown in Fig. 6A. The areas illuminated by GFP expression with the Dino-Lite digital microscope were resected as recurrent tumors. A representative image of the FGS + PIT group 4 weeks after surgery is shown in Fig. 6B. The harvested tumor showed invasion into the muscles and was mainly fibrostic with extensive stroma (Fig. 6C). A comparison of the perioperative data between the two groups is presented in Table 1. The local recurrence rate was 100.0% for the FGS-only group and 50.0% for the FGS + PIT group. The metastatic recurrence rate was 100.0% for the FGS-only group and 25.0% for the FGS + PIT group, being significantly less frequent in the latter. The total weight of recurrent tumors in the FGS +PIT group was significantly lower than that in the FGS group (2370.3 ± 1907.8 versus 705.5 ± 1200.0 mg, p = 0.039).
Table 1
Comparison of perioperative outcomes between FGS-only and FGS combined with PIT
 
FGS-only
(n = 8)
FGS + PIT
(n = 8)
p-value
Body weight (g)
26.4 ± 3.0
26.8 ± 2.8
0.65
Tumor-to-pancreas ratio
3.60 ± 0.38
3.83 ± 0.83
0.51
Tumor volume (mm3)
211.3 ± 48.4
207.3 ± 95.6
0.57
Specimen weight (mg)
316 ± 109
260 ± 170
0.19
Local recurrence rate (%)
100.0%
50.0%
0.077
Metastatic recurrence rate (%)
100.0%
25.0%
0.0070
Total recurrent tumor weight (mg)
2370.3 ± 1907.8
705.5 ± 1200.0
0.039

Discussion

In the present study, we demonstrated that the combination of FGS and adjuvant PIT, using a single anti-CEACAM antibody with a 700-nm fluorophore conjugate, could reduce recurrences in an orthotopic mouse model of pancreatic cancer.
Medical devices for FGS used in clinical practice are mainly 800 nm for ICG. Our laboratory has reported colon or pancreatic cancer labeling using tumor-specific antibodies conjugated to a 800-nm fluorescent dye.7,20,21 Anti-CEACAM has been shown to be useful for detection of colon cancer at both 800 and 700 nm in mouse models.14,17 Although 700 nm is less penetrant than 800 nm, the development of medical devices for FGS using 700 nm has been progressing. The introduction of FGS using a 700-nm fluorescent dye is promising for the advancement of color-coded surgery, in which organs are fluorescently labeled at various wavelengths.
PIT is a molecularly targeted phototherapy for cancer that is based on injecting a conjugate of a near-infrared IR700DX and a monoclonal antibody that targets an antigen expressed on the cancer cell surface. Subsequent local exposure to NIR light results in rapid and highly selective immunogenic death of targeted cancer cells.9 NIR-PIT not only treats the targeted local tumor but also reduces or eliminates systemic metastasis and prevents recurrence in animal models.22, 23 In the present study, PIT performed as an intraoperative adjuvant therapy reduced postoperative recurrence in an orthotopic mouse model of pancreatic cancer.
Our previous report clarified that 6G5j can bind to CEACAM1, CEACAM3, CEACAM5 (conventional CEA), CEACAM6, and CEACAM8.17 The ability to bind to multiple CEACAMs may improve the binding capacity of the antibody and enhance the visualization of tumor margins that are otherwise invisible compared with CEA alone.
Regarding the association between CEACAM and pancreatic cancer, approximately 70% of all analyzed tumor tissues showed expression of CEACAM 1, 5, or 6, or a combination of all of them.24 CEACAM 5 and 6 expression were correlated with lymph node metastasis, and patients with high expression of CEACAM 5 or 6 had shorter overall survival and disease-free survival.24 A CEACAM antibody is expected to be useful in the targeting of pancreatic cancer, and the present study demonstrated this in a BxPC-3-GFP pancreatic cancer mouse model.
This is the first report of combined FGS and PIT using a single fluorescent antibody. If multiple agents are used in combination, there could be concern about their interactions; however, a single compound could reduce the likelihood of this happening.
The present proof-of-principle study demonstrates that FGS and intraoperative adjuvant PIT can be combined by using 6G5j-IR700DX in an orthotopic mouse model of the human BxPC-3-GFP pancreatic cancer cell line with the combination contributing to the inhibition of tumor recurrence. Currently, pancreatic cancer located in the uncinate process has a higher rate of R1 resection than nonuncinate process pancreatic head cancers. In addition, survival rates are lower after R0 resection of uncinate process pancreatic cancer.25 To improve outcomes in this recalcitrant cancer, we will first develop, in future experiments, an orthotopic model of uncinate cancer to determine whether FGS can improve outcomes. Secondly, we will develop the orthotopic uncinate process model in humanized NSG mice engrafted with peripheral blood lymphocytes.26 We will combine FGS and photoimmunotherapy (PIT) on the orthotopic uncinate process cancer in the humanized model to determine the advantage of the combined modalities.

Conclusions

FGS and adjuvant PIT can be combined by using a single anti-CEACAM antibody with 700-nm fluorophore conjugate in an orthotopic mouse model of pancreatic cancer. Adjuvant intraoperative PIT after FGS could significantly reduce the frequency of recurrence and the total weight of recurrent tumors, which suggests clinical potential of combining FGS with PIT.

Acknowledgments

This study was funded by Veterans Affairs Merit Review grant nos. 1 I01 BX003856-01A1 and 1 I01 BX004494-01 (M.B.), NIH/NCI R01CA256973 (M.B.), NIH/NCI T32CA121938 (M.A.T. and T.M.L.), and The Uehara Memorial Foundation (H.N.).
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33.CrossRef Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33.CrossRef
2.
Zurück zum Zitat Sirri E, Castro FA, Kieschke J, Jansen L, Emrich K, Gondos A, et al. Recent trends in survival of patients with pancreatic cancer in Germany and the United States. Pancreas. 2016;45(6):908–14.CrossRef Sirri E, Castro FA, Kieschke J, Jansen L, Emrich K, Gondos A, et al. Recent trends in survival of patients with pancreatic cancer in Germany and the United States. Pancreas. 2016;45(6):908–14.CrossRef
3.
Zurück zum Zitat Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.CrossRef Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.CrossRef
4.
Zurück zum Zitat Lwin TM, Hoffman RM, Bouvet M. The development of fluorescence guided surgery for pancreatic cancer: from bench to clinic. Expert Rev Anticancer Ther. 2018;18(7):651–62.CrossRef Lwin TM, Hoffman RM, Bouvet M. The development of fluorescence guided surgery for pancreatic cancer: from bench to clinic. Expert Rev Anticancer Ther. 2018;18(7):651–62.CrossRef
5.
Zurück zum Zitat Gutowski M, Framery B, Boonstra MC, Garambois V, Quenet F, Dumas K, et al. SGM-101: an innovative near-infrared dye-antibody conjugate that targets CEA for fluorescence-guided surgery. Surg Oncol. 2017;26:153–62.CrossRef Gutowski M, Framery B, Boonstra MC, Garambois V, Quenet F, Dumas K, et al. SGM-101: an innovative near-infrared dye-antibody conjugate that targets CEA for fluorescence-guided surgery. Surg Oncol. 2017;26:153–62.CrossRef
6.
Zurück zum Zitat Hiroshima Y, Maawy A, Zhang Y, Murakami T, Momiyama M, Mori R, et al. Fluorescence-guided surgery, but not bright-light surgery, prevents local recurrence in a pancreatic cancer patient derived orthotopic xenograft (PDOX) model resistant to neoadjuvant chemotherapy (NAC). Pancreatology. 2015;15:295–301.CrossRef Hiroshima Y, Maawy A, Zhang Y, Murakami T, Momiyama M, Mori R, et al. Fluorescence-guided surgery, but not bright-light surgery, prevents local recurrence in a pancreatic cancer patient derived orthotopic xenograft (PDOX) model resistant to neoadjuvant chemotherapy (NAC). Pancreatology. 2015;15:295–301.CrossRef
7.
Zurück zum Zitat Lwin TM, Murakami T, Miyake K, Yazaki PJ, Shivley JE, Hoffman RM, et al. Tumor-specific labeling of pancreatic cancer using a humanized anti-CEA antibody conjugated to a near-infrared fluorophore. Ann Surg Oncol. 2018;25:1079–85.CrossRef Lwin TM, Murakami T, Miyake K, Yazaki PJ, Shivley JE, Hoffman RM, et al. Tumor-specific labeling of pancreatic cancer using a humanized anti-CEA antibody conjugated to a near-infrared fluorophore. Ann Surg Oncol. 2018;25:1079–85.CrossRef
8.
Zurück zum Zitat Mitsunaga M, Ogawa M, Kosaka N, Rosenblum LT, Choyke PL, Kobayashi H. Cancer cell-selective in vivo near infrared photoimmunotherapy targeting specific membrane molecules. Nat Med. 2011;17:1685–91.CrossRef Mitsunaga M, Ogawa M, Kosaka N, Rosenblum LT, Choyke PL, Kobayashi H. Cancer cell-selective in vivo near infrared photoimmunotherapy targeting specific membrane molecules. Nat Med. 2011;17:1685–91.CrossRef
9.
Zurück zum Zitat Kobayashi H, Choyke PL. Near-infrared photoimmunotherapy of cancer. Acc Chem Res. 2019;52:2332–9.CrossRef Kobayashi H, Choyke PL. Near-infrared photoimmunotherapy of cancer. Acc Chem Res. 2019;52:2332–9.CrossRef
10.
Zurück zum Zitat Maawy AA, Hiroshima Y, Zhang Y, Heim R, Makings L, Garcia-Guzman M, et al. Near infra-red photoimmunotherapy with anti-CEA-IR700 results in extensive tumor lysis and a significant decrease in tumor burden in orthotopic mouse models of pancreatic cancer. PLoS One. 2015;10:e0121989.CrossRef Maawy AA, Hiroshima Y, Zhang Y, Heim R, Makings L, Garcia-Guzman M, et al. Near infra-red photoimmunotherapy with anti-CEA-IR700 results in extensive tumor lysis and a significant decrease in tumor burden in orthotopic mouse models of pancreatic cancer. PLoS One. 2015;10:e0121989.CrossRef
11.
Zurück zum Zitat Maawy AA, Hiroshima Y, Zhang Y, Garcia-Guzman M, Luiken GA, Kobayashi H, et al. Photoimmunotherapy lowers recurrence after pancreatic cancer surgery in orthotopic nude mouse models. J Surg Res. 2015;197:5–11.CrossRef Maawy AA, Hiroshima Y, Zhang Y, Garcia-Guzman M, Luiken GA, Kobayashi H, et al. Photoimmunotherapy lowers recurrence after pancreatic cancer surgery in orthotopic nude mouse models. J Surg Res. 2015;197:5–11.CrossRef
12.
Zurück zum Zitat Hiroshima Y, Maawy A, Zhang Y, Guzman MG, Heim R, Makings L, et al. Photoimmunotherapy inhibits tumor recurrence after surgical resection on a pancreatic cancer patient-derived orthotopic xenograft (PDOX) nude mouse model. Ann Surg Oncol. 2015;22:S1469-1474.CrossRef Hiroshima Y, Maawy A, Zhang Y, Guzman MG, Heim R, Makings L, et al. Photoimmunotherapy inhibits tumor recurrence after surgical resection on a pancreatic cancer patient-derived orthotopic xenograft (PDOX) nude mouse model. Ann Surg Oncol. 2015;22:S1469-1474.CrossRef
13.
Zurück zum Zitat Beauchemin N, Arabzadeh A. Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) in cancer progression and metastasis. Cancer Metastasis Rev. 2013;32:643–71.CrossRef Beauchemin N, Arabzadeh A. Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) in cancer progression and metastasis. Cancer Metastasis Rev. 2013;32:643–71.CrossRef
14.
Zurück zum Zitat Nishino H, Hollandsworth HM, Amirfakhri S, Tashiro Y, Yamamoto J, Turner MA, et al. A Novel color-coded liver metastasis mouse model to distinguish tumor and adjacent liver segment. J Surg Res. 2021;264:327–33.CrossRef Nishino H, Hollandsworth HM, Amirfakhri S, Tashiro Y, Yamamoto J, Turner MA, et al. A Novel color-coded liver metastasis mouse model to distinguish tumor and adjacent liver segment. J Surg Res. 2021;264:327–33.CrossRef
15.
Zurück zum Zitat Fu X, Guadagni F, Hoffman RM. A metastatic nude-mouse model of human pancreatic cancer constructed orthotopically with histologically intact patient specimens. Proc Natl Acad Sci. 1992;89:5645–9.CrossRef Fu X, Guadagni F, Hoffman RM. A metastatic nude-mouse model of human pancreatic cancer constructed orthotopically with histologically intact patient specimens. Proc Natl Acad Sci. 1992;89:5645–9.CrossRef
16.
Zurück zum Zitat Furukawa T, Kubota T, Watanabe M, Kitajima M, Hoffman RM. A novel “patient-like” treatment model of human pancreatic cancer constructed using orthotopic transplantation of histologically intact human tumor tissue in nude mice. Cancer Res. 1993;53:3070–2. Furukawa T, Kubota T, Watanabe M, Kitajima M, Hoffman RM. A novel “patient-like” treatment model of human pancreatic cancer constructed using orthotopic transplantation of histologically intact human tumor tissue in nude mice. Cancer Res. 1993;53:3070–2.
17.
Zurück zum Zitat Hollandsworth HM, Amirfakhri S, Filemoni F, Schmitt V, Wennemuth G, Schmidt A, et al. Anti-carcinoembryonic antigen-related cell adhesion molecule antibody for fluorescence visualization of primary colon cancer and metastases in patient-derived orthotopic xenograft mouse models. Oncotarget. 2020;11:429–39.CrossRef Hollandsworth HM, Amirfakhri S, Filemoni F, Schmitt V, Wennemuth G, Schmidt A, et al. Anti-carcinoembryonic antigen-related cell adhesion molecule antibody for fluorescence visualization of primary colon cancer and metastases in patient-derived orthotopic xenograft mouse models. Oncotarget. 2020;11:429–39.CrossRef
18.
Zurück zum Zitat Hollandsworth HM, Amirfakhri S, Filemoni F, Hoffman RM, Molnar J, Yazaki PJ, et al. Humanized anti-tumor-associated glycoprotein-72 for submillimeter near-infrared detection of colon cancer in metastatic mouse models. J Surg Res. 2020;252:16–21.CrossRef Hollandsworth HM, Amirfakhri S, Filemoni F, Hoffman RM, Molnar J, Yazaki PJ, et al. Humanized anti-tumor-associated glycoprotein-72 for submillimeter near-infrared detection of colon cancer in metastatic mouse models. J Surg Res. 2020;252:16–21.CrossRef
19.
Zurück zum Zitat Hollandsworth HM, Amirfakhri S, Filemoni F, Molnar J, Hoffman RM, Yazaki P, et al. Near-infrared photoimmunotherapy is effective treatment for colorectal cancer in orthotopic nude-mouse models. PLoS One. 2020;15:e0234643.CrossRef Hollandsworth HM, Amirfakhri S, Filemoni F, Molnar J, Hoffman RM, Yazaki P, et al. Near-infrared photoimmunotherapy is effective treatment for colorectal cancer in orthotopic nude-mouse models. PLoS One. 2020;15:e0234643.CrossRef
20.
Zurück zum Zitat Lwin TM, Hernot S, Hollandsworth H, Amirfakhri S, Filemoni F, Debie P, et al. Tumor-specific near-infrared nanobody probe rapidly labels tumors in an orthotopic mouse model of pancreatic cancer. Surgery. 2020;168:85–91.CrossRef Lwin TM, Hernot S, Hollandsworth H, Amirfakhri S, Filemoni F, Debie P, et al. Tumor-specific near-infrared nanobody probe rapidly labels tumors in an orthotopic mouse model of pancreatic cancer. Surgery. 2020;168:85–91.CrossRef
21.
Zurück zum Zitat Hollandsworth HM, Schmitt V, Amirfakhri S, Filemoni F, Schmidt A, Landström M, et al. Fluorophore-conjugated Helicobacter pylori recombinant membrane protein (HopQ) labels primary colon cancer and metastases in orthotopic mouse models by binding CEA-related cell adhesion molecules. Transl Oncol. 2020;13:100857.CrossRef Hollandsworth HM, Schmitt V, Amirfakhri S, Filemoni F, Schmidt A, Landström M, et al. Fluorophore-conjugated Helicobacter pylori recombinant membrane protein (HopQ) labels primary colon cancer and metastases in orthotopic mouse models by binding CEA-related cell adhesion molecules. Transl Oncol. 2020;13:100857.CrossRef
22.
Zurück zum Zitat Sato K, Nagaya T, Nakamura Y, Harada T, Choyke PL, Kobayashi H. Near infrared photoimmunotherapy prevents lung cancer metastases in a murine model. Oncotarget. 2015;6:19747–58.CrossRef Sato K, Nagaya T, Nakamura Y, Harada T, Choyke PL, Kobayashi H. Near infrared photoimmunotherapy prevents lung cancer metastases in a murine model. Oncotarget. 2015;6:19747–58.CrossRef
23.
Zurück zum Zitat Sato K, Nagaya T, Mitsunaga M, Choyke PL, Kobayashi H. Near infrared photoimmunotherapy for lung metastases. Cancer Lett. 2015;365:112–21.CrossRef Sato K, Nagaya T, Mitsunaga M, Choyke PL, Kobayashi H. Near infrared photoimmunotherapy for lung metastases. Cancer Lett. 2015;365:112–21.CrossRef
24.
Zurück zum Zitat Gebauer F, Wicklein D, Horst J, Sundermann P, Maar H, Streichert T, et al. Carcinoembryonic antigen-related cell adhesion molecules (CEACAM) 1, 5 and 6 as biomarkers in pancreatic cancer. PLoS ONE. 2014;9:e113023.CrossRef Gebauer F, Wicklein D, Horst J, Sundermann P, Maar H, Streichert T, et al. Carcinoembryonic antigen-related cell adhesion molecules (CEACAM) 1, 5 and 6 as biomarkers in pancreatic cancer. PLoS ONE. 2014;9:e113023.CrossRef
25.
Zurück zum Zitat Liu C, Tian X, Xie X, Gao H, Zhuang Y, et al. Comparison of uncinate process cancer and non-uncinate process pancreatic head cancers. J Cancer. 2016;7:1242–9.CrossRef Liu C, Tian X, Xie X, Gao H, Zhuang Y, et al. Comparison of uncinate process cancer and non-uncinate process pancreatic head cancers. J Cancer. 2016;7:1242–9.CrossRef
26.
Zurück zum Zitat Abate-Daga D, Lagisetty KH, Tran E, Zheng Z, Gattinoti L, et al. A novel chimeric antigen receptor against prostate stem cell antigen mediates tumor destruction in a humanized mouse model of pancreatic cancer. Hum Gene Ther. 2014;25:1003–12.CrossRef Abate-Daga D, Lagisetty KH, Tran E, Zheng Z, Gattinoti L, et al. A novel chimeric antigen receptor against prostate stem cell antigen mediates tumor destruction in a humanized mouse model of pancreatic cancer. Hum Gene Ther. 2014;25:1003–12.CrossRef
Metadaten
Titel
Proof of Principle of Combining Fluorescence-Guided Surgery with Photoimmunotherapy to Improve the Outcome of Pancreatic Cancer Therapy in an Orthotopic Mouse Model
verfasst von
Hiroto Nishino, MD, PhD
Michael A. Turner, MD
Siamak Amirfakhri, PhD, DVM
Thinzar M. Lwin, MD, MS
Mojgan Hosseini, MD
Bernhard B. Singer, PhD
Robert M. Hoffman, PhD
Michael Bouvet, MD
Publikationsdatum
04.09.2022
Verlag
Springer International Publishing
Erschienen in
Annals of Surgical Oncology / Ausgabe 1/2023
Print ISSN: 1068-9265
Elektronische ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-022-12466-4

Weitere Artikel der Ausgabe 1/2023

Annals of Surgical Oncology 1/2023 Zur Ausgabe

Fehlerkultur in der Medizin – Offenheit zählt!

Darüber reden und aus Fehlern lernen, sollte das Motto in der Medizin lauten. Und zwar nicht nur im Sinne der Patientensicherheit. Eine negative Fehlerkultur kann auch die Behandelnden ernsthaft krank machen, warnt Prof. Dr. Reinhard Strametz. Ein Plädoyer und ein Leitfaden für den offenen Umgang mit kritischen Ereignissen in Medizin und Pflege.

Mehr Frauen im OP – weniger postoperative Komplikationen

21.05.2024 Allgemeine Chirurgie Nachrichten

Ein Frauenanteil von mindestens einem Drittel im ärztlichen Op.-Team war in einer großen retrospektiven Studie aus Kanada mit einer signifikanten Reduktion der postoperativen Morbidität assoziiert.

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.