Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 2/2018

01-02-2018 | Original Article

Cholecystokinin receptor antagonist alters pancreatic cancer microenvironment and increases efficacy of immune checkpoint antibody therapy in mice

Authors: Jill P. Smith, Shangzi Wang, Sandeep Nadella, Sandra A. Jablonski, Louis M. Weiner

Published in: Cancer Immunology, Immunotherapy | Issue 2/2018

Login to get access

Abstract

Advanced pancreatic ductal adenocarcinoma (PDAC) has typically been resistant to chemotherapy and immunotherapy; therefore, novel strategies are needed to enhance therapeutic response. Cholecystokinin (CCK) has been shown to stimulate growth of pancreatic cancer. CCK receptors (CCKRs) are present on pancreatic cancer cells, fibroblasts, and lymphocytes. We hypothesized that CCKR blockade would improve response to immune checkpoint antibodies by promoting influx of tumor-infiltrating lymphocytes (TILs) and reducing fibrosis. We examined the effects of CCKR antagonists or immune checkpoint blockade antibodies alone or in combination in murine models of PDAC. Monotherapy with CCKR blockade significantly decreased tumor size and metastases in SCID mice with orthotopic PDAC, and in C57BL/6 mice, it reduced fibrosis and induced the influx of TILs. Immune-competent mice bearing syngeneic pancreatic cancer (Panc02 and mT3-2D) that were treated with the combination of CCK receptor antagonists and immune checkpoint blockade antibodies survived significantly longer with smaller tumors. Tumor immunohistochemical staining and flow cytometry demonstrated that the tumors of mice treated with the combination regimen had a significant reduction in Foxp3+ T-regulatory cells and an increase in CD4+ and CD8+ lymphocytes. Masson’s trichrome stain analysis revealed 50% less fibrosis in the tumors of mice treated with CCKR antagonist compared to controls and compared to checkpoint antibody therapy. CCKR antagonists given with immune checkpoint antibody therapy represent a novel approach for improving survival of PDAC. The mechanism by which this combination therapy improves the survival of PDAC may be related to the decreased fibrosis and immune cells of the tumor microenvironment.
Appendix
Available only for authorised users
Literature
1.
go back to reference Smith JP, Solomon TE, Bagheri S, Kramer S (1990) Cholecystokinin stimulates growth of human pancreatic adenocarcinoma SW-1990. Dig Dis Sci 35:1377–1384CrossRefPubMed Smith JP, Solomon TE, Bagheri S, Kramer S (1990) Cholecystokinin stimulates growth of human pancreatic adenocarcinoma SW-1990. Dig Dis Sci 35:1377–1384CrossRefPubMed
2.
go back to reference Smith JP, Kramer ST, Solomon TE (1991) CCK stimulates growth of six human pancreatic cancer cell lines in serum-free medium. Regul Pept 32:341–349CrossRefPubMed Smith JP, Kramer ST, Solomon TE (1991) CCK stimulates growth of six human pancreatic cancer cell lines in serum-free medium. Regul Pept 32:341–349CrossRefPubMed
3.
go back to reference Smith JP, Fantaskey AP, Liu G, Zagon IS (1995) Identification of gastrin as a growth peptide in human pancreatic cancer. Am J Physiol 268:R135–R141PubMed Smith JP, Fantaskey AP, Liu G, Zagon IS (1995) Identification of gastrin as a growth peptide in human pancreatic cancer. Am J Physiol 268:R135–R141PubMed
4.
7.
go back to reference Brand SJ, Fuller PJ (1988) Differential gastrin gene expression in rat gastrointestinal tract and pancreas during neonatal development. J Biol Chem 263:5341–5347PubMed Brand SJ, Fuller PJ (1988) Differential gastrin gene expression in rat gastrointestinal tract and pancreas during neonatal development. J Biol Chem 263:5341–5347PubMed
8.
go back to reference Tamiolakis D, Venizelos I, Simopoulos C, Kotini A, Jivannakis T, Papadopoulos N (2004) Does neoplastic gastrin expression remodel the embryonal pattern of the protein? A study in human pancreas. Hepatogastroenterology 51:249–252PubMed Tamiolakis D, Venizelos I, Simopoulos C, Kotini A, Jivannakis T, Papadopoulos N (2004) Does neoplastic gastrin expression remodel the embryonal pattern of the protein? A study in human pancreas. Hepatogastroenterology 51:249–252PubMed
9.
go back to reference Prasad NB, Biankin AV, Fukushima N, Maitra A, Dhara S, Elkahloun AG, Hruban RH, Goggins M, Leach SD (2005) Gene expression profiles in pancreatic intraepithelial neoplasia reflect the effects of Hedgehog signaling on pancreatic ductal epithelial cells. Cancer Res 65:1619–1626. doi:10.1158/0008-5472.CAN-04-1413 CrossRefPubMed Prasad NB, Biankin AV, Fukushima N, Maitra A, Dhara S, Elkahloun AG, Hruban RH, Goggins M, Leach SD (2005) Gene expression profiles in pancreatic intraepithelial neoplasia reflect the effects of Hedgehog signaling on pancreatic ductal epithelial cells. Cancer Res 65:1619–1626. doi:10.​1158/​0008-5472.​CAN-04-1413 CrossRefPubMed
10.
go back to reference Smith JP, Hamory MW, Verderame MF, Zagon IS (1998) Quantitative analysis of gastrin mRNA and peptide in normal and cancerous human pancreas. Int J Mol Med 2:309–315PubMed Smith JP, Hamory MW, Verderame MF, Zagon IS (1998) Quantitative analysis of gastrin mRNA and peptide in normal and cancerous human pancreas. Int J Mol Med 2:309–315PubMed
11.
go back to reference Smith JP, Shih A, Wu Y, McLaughlin PJ, Zagon IS (1996) Gastrin regulates growth of human pancreatic cancer in a tonic and autocrine fashion. Am J Physiol 270:R1078–R1084CrossRefPubMed Smith JP, Shih A, Wu Y, McLaughlin PJ, Zagon IS (1996) Gastrin regulates growth of human pancreatic cancer in a tonic and autocrine fashion. Am J Physiol 270:R1078–R1084CrossRefPubMed
12.
go back to reference Smith JP, Rickabaugh CA, McLaughlin PJ, Zagon IS (1993) Cholecystokinin receptors and PANC-1 human pancreatic cancer cells. Am J Physiol 265:G149–G155PubMed Smith JP, Rickabaugh CA, McLaughlin PJ, Zagon IS (1993) Cholecystokinin receptors and PANC-1 human pancreatic cancer cells. Am J Physiol 265:G149–G155PubMed
13.
go back to reference Smith JP, Liu G, Soundararajan V, McLaughlin PJ, Zagon IS (1994) Identification and characterization of CCK-B/gastrin receptors in human pancreatic cancer cell lines. Am J Physiol 266:R277–R283PubMed Smith JP, Liu G, Soundararajan V, McLaughlin PJ, Zagon IS (1994) Identification and characterization of CCK-B/gastrin receptors in human pancreatic cancer cell lines. Am J Physiol 266:R277–R283PubMed
14.
go back to reference Matters GL, Harms JF, McGovern CO, Jayakumar C, Crepin K, Smith ZP, Nelson MC, Stock H, Fenn CW, Kaiser J, Kester M, Smith JP (2009) Growth of human pancreatic cancer is inhibited by down-regulation of gastrin gene expression. Pancreas 38:e151–e161CrossRefPubMedPubMedCentral Matters GL, Harms JF, McGovern CO, Jayakumar C, Crepin K, Smith ZP, Nelson MC, Stock H, Fenn CW, Kaiser J, Kester M, Smith JP (2009) Growth of human pancreatic cancer is inhibited by down-regulation of gastrin gene expression. Pancreas 38:e151–e161CrossRefPubMedPubMedCentral
17.
go back to reference Wank SA, Harkins R, Jensen RT, Shapira H, de Weerth A, Slattery T (1992) Purification, molecular cloning, and functional expression of the cholecystokinin receptor from rat pancreas. Proc Natl Acad Sci USA 89:3125–3129CrossRefPubMedPubMedCentral Wank SA, Harkins R, Jensen RT, Shapira H, de Weerth A, Slattery T (1992) Purification, molecular cloning, and functional expression of the cholecystokinin receptor from rat pancreas. Proc Natl Acad Sci USA 89:3125–3129CrossRefPubMedPubMedCentral
18.
go back to reference Wank SA, Pisegna JR, de Weerth A (1994) Cholecystokinin receptor family. Molecular cloning, structure, and functional expression in rat, guinea pig, and human. Ann N Y Acad Sci 713:49–66CrossRefPubMed Wank SA, Pisegna JR, de Weerth A (1994) Cholecystokinin receptor family. Molecular cloning, structure, and functional expression in rat, guinea pig, and human. Ann N Y Acad Sci 713:49–66CrossRefPubMed
19.
go back to reference Weinberg DS, Ruggeri B, Barber MT, Biswas S, Miknyocki S, Waldman SA (1997) Cholecystokinin A and B receptors are differentially expressed in normal pancreas and pancreatic adenocarcinoma. J Clin Invest 100:597–603CrossRefPubMedPubMedCentral Weinberg DS, Ruggeri B, Barber MT, Biswas S, Miknyocki S, Waldman SA (1997) Cholecystokinin A and B receptors are differentially expressed in normal pancreas and pancreatic adenocarcinoma. J Clin Invest 100:597–603CrossRefPubMedPubMedCentral
20.
go back to reference Longnecker DS, Curphey TJ, Lilja HS, French JI, Daniel DS (1980) Carcinogenicity in rats of the nitrosourea amino acid N delta-(N-methyl-N-nitrosocarbamoyl)-L-ornithine. J Environ Pathol Toxicol 4:117–129PubMed Longnecker DS, Curphey TJ, Lilja HS, French JI, Daniel DS (1980) Carcinogenicity in rats of the nitrosourea amino acid N delta-(N-methyl-N-nitrosocarbamoyl)-L-ornithine. J Environ Pathol Toxicol 4:117–129PubMed
22.
go back to reference Smith JP, Verderame MF, McLaughlin P, Martenis M, Ballard E, Zagon IS (2002) Characterization of the CCK-C (cancer) receptor in human pancreatic cancer. Int J Mol Med 10:689–694PubMed Smith JP, Verderame MF, McLaughlin P, Martenis M, Ballard E, Zagon IS (2002) Characterization of the CCK-C (cancer) receptor in human pancreatic cancer. Int J Mol Med 10:689–694PubMed
23.
24.
25.
go back to reference Abbruzzese JL, Gholson CF, Daugherty K, Larson E, DuBrow R, Berlin R, Levin B (1992) A pilot clinical trial of the cholecystokinin receptor antagonist MK-329 in patients with advanced pancreatic cancer. Pancreas 7:165–171CrossRefPubMed Abbruzzese JL, Gholson CF, Daugherty K, Larson E, DuBrow R, Berlin R, Levin B (1992) A pilot clinical trial of the cholecystokinin receptor antagonist MK-329 in patients with advanced pancreatic cancer. Pancreas 7:165–171CrossRefPubMed
26.
go back to reference Berna MJ, Jensen RT (2007) Role of CCK/gastrin receptors in gastrointestinal/metabolic diseases and results of human studies using gastrin/CCK receptor agonists/antagonists in these diseases. Curr Top Med Chem 7:1211–1231CrossRefPubMedPubMedCentral Berna MJ, Jensen RT (2007) Role of CCK/gastrin receptors in gastrointestinal/metabolic diseases and results of human studies using gastrin/CCK receptor agonists/antagonists in these diseases. Curr Top Med Chem 7:1211–1231CrossRefPubMedPubMedCentral
27.
go back to reference Chang RS, Lotti VJ, Chen TB, Kunkel KA (1986) Characterization of the binding of [3H]-(±)-L-364,718: a new potent, nonpeptide cholecystokinin antagonist radioligand selective for peripheral receptors. Mol Pharmacol 30:212–217PubMed Chang RS, Lotti VJ, Chen TB, Kunkel KA (1986) Characterization of the binding of [3H]-(±)-L-364,718: a new potent, nonpeptide cholecystokinin antagonist radioligand selective for peripheral receptors. Mol Pharmacol 30:212–217PubMed
29.
go back to reference Hahne WF, Jensen RT, Lemp GF, Gardner JD (1981) Proglumide and benzotript: members of a different class of cholecystokinin receptor antagonists. Proc Natl Acad Sci USA 78:6304–6308CrossRefPubMedPubMedCentral Hahne WF, Jensen RT, Lemp GF, Gardner JD (1981) Proglumide and benzotript: members of a different class of cholecystokinin receptor antagonists. Proc Natl Acad Sci USA 78:6304–6308CrossRefPubMedPubMedCentral
30.
go back to reference Singh P, Owlia A, Espeijo R, Dai B (1995) Novel gastrin receptors mediate mitogenic effects of gastrin and processing intermediates of gastrin on Swiss 3T3 fibroblasts. Absence of detectable cholecystokinin (CCK)-A and CCK-B receptors. J Biol Chem 270:8429–8438CrossRefPubMed Singh P, Owlia A, Espeijo R, Dai B (1995) Novel gastrin receptors mediate mitogenic effects of gastrin and processing intermediates of gastrin on Swiss 3T3 fibroblasts. Absence of detectable cholecystokinin (CCK)-A and CCK-B receptors. J Biol Chem 270:8429–8438CrossRefPubMed
32.
go back to reference Phillips PA, Yang L, Shulkes A, Vonlaufen A, Poljak A, Bustamante S, Warren A, Xu Z, Guilhaus M, Pirola R, Apte MV, Wilson JS (2010) Pancreatic stellate cells produce acetylcholine and may play a role in pancreatic exocrine secretion. Proc Natl Acad Sci USA 107:17397–17402. doi:10.1073/pnas.1000359107 CrossRefPubMedPubMedCentral Phillips PA, Yang L, Shulkes A, Vonlaufen A, Poljak A, Bustamante S, Warren A, Xu Z, Guilhaus M, Pirola R, Apte MV, Wilson JS (2010) Pancreatic stellate cells produce acetylcholine and may play a role in pancreatic exocrine secretion. Proc Natl Acad Sci USA 107:17397–17402. doi:10.​1073/​pnas.​1000359107 CrossRefPubMedPubMedCentral
33.
go back to reference Apte MV, Park S, Phillips PA, Santucci N, Goldstein D, Kumar RK, Ramm GA, Buchler M, Friess H, McCarroll JA, Keogh G, Merrett N, Pirola R, Wilson JS (2004) Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas 29:179–187CrossRefPubMed Apte MV, Park S, Phillips PA, Santucci N, Goldstein D, Kumar RK, Ramm GA, Buchler M, Friess H, McCarroll JA, Keogh G, Merrett N, Pirola R, Wilson JS (2004) Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas 29:179–187CrossRefPubMed
36.
go back to reference Zhang JG, Cong B, Li QX, Chen HY, Qin J, Fu LH (2011) Cholecystokinin octapeptide regulates lipopolysaccharide-activated B cells co-stimulatory molecule expression and cytokines production in vitro. Immunopharmacol Immunotoxicol 33:157–163. doi:10.3109/08923973.2010.491079 CrossRefPubMed Zhang JG, Cong B, Li QX, Chen HY, Qin J, Fu LH (2011) Cholecystokinin octapeptide regulates lipopolysaccharide-activated B cells co-stimulatory molecule expression and cytokines production in vitro. Immunopharmacol Immunotoxicol 33:157–163. doi:10.​3109/​08923973.​2010.​491079 CrossRefPubMed
37.
go back to reference Corbett TH, Roberts BJ, Leopold WR, Peckham JC, Wilkoff LJ, Griswold DP Jr, Schabel FM Jr (1984) Induction and chemotherapeutic response of two transplantable ductal adenocarcinomas of the pancreas in C57BL/6 mice. Cancer Res 44:717–726PubMed Corbett TH, Roberts BJ, Leopold WR, Peckham JC, Wilkoff LJ, Griswold DP Jr, Schabel FM Jr (1984) Induction and chemotherapeutic response of two transplantable ductal adenocarcinomas of the pancreas in C57BL/6 mice. Cancer Res 44:717–726PubMed
38.
go back to reference Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, Jager M, Ponz-Sarvise M, Tiriac H, Spector MS, Gracanin A, Oni T, Yu KH, van BR, Huch M, Rivera KD, Wilson JP, Feigin ME, Ohlund D, Handly-Santana A, Ardito-Abraham CM, Ludwig M, Elyada E, Alagesan B, Biffi G, Yordanov GN, Delcuze B, Creighton B, Wright K, Park Y, Morsink FH, Molenaar IQ, Borel Rinkes IH, Cuppen E, Hao Y, Jin Y, Nijman IJ, Iacobuzio-Donahue C, Leach SD, Pappin DJ, Hammell M, Klimstra DS, Basturk O, Hruban RH, Offerhaus GJ, Vries RG, Clevers H, Tuveson DA (2015) Organoid models of human and mouse ductal pancreatic cancer. Cell 160:324–338. doi:10.1016/j.cell.2014.12.021 CrossRefPubMed Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, Jager M, Ponz-Sarvise M, Tiriac H, Spector MS, Gracanin A, Oni T, Yu KH, van BR, Huch M, Rivera KD, Wilson JP, Feigin ME, Ohlund D, Handly-Santana A, Ardito-Abraham CM, Ludwig M, Elyada E, Alagesan B, Biffi G, Yordanov GN, Delcuze B, Creighton B, Wright K, Park Y, Morsink FH, Molenaar IQ, Borel Rinkes IH, Cuppen E, Hao Y, Jin Y, Nijman IJ, Iacobuzio-Donahue C, Leach SD, Pappin DJ, Hammell M, Klimstra DS, Basturk O, Hruban RH, Offerhaus GJ, Vries RG, Clevers H, Tuveson DA (2015) Organoid models of human and mouse ductal pancreatic cancer. Cell 160:324–338. doi:10.​1016/​j.​cell.​2014.​12.​021 CrossRefPubMed
40.
go back to reference Kennedy DJ, Vetteth S, Periyasamy SM, Kanj M, Fedorova L, Khouri S, Kahaleh MB, Xie Z, Malhotra D, Kolodkin NI, Lakatta EG, Fedorova OV, Bagrov AY, Shapiro JI (2006) Central role for the cardiotonic steroid marinobufagenin in the pathogenesis of experimental uremic cardiomyopathy. Hypertension 47:488–495. doi:10.1161/01.HYP.0000202594.82271.92 CrossRefPubMed Kennedy DJ, Vetteth S, Periyasamy SM, Kanj M, Fedorova L, Khouri S, Kahaleh MB, Xie Z, Malhotra D, Kolodkin NI, Lakatta EG, Fedorova OV, Bagrov AY, Shapiro JI (2006) Central role for the cardiotonic steroid marinobufagenin in the pathogenesis of experimental uremic cardiomyopathy. Hypertension 47:488–495. doi:10.​1161/​01.​HYP.​0000202594.​82271.​92 CrossRefPubMed
42.
go back to reference Hanyu N, Dodds WJ, Layman RD, Hogan WJ, Colton DG (1991) Effect of two new cholecystokinin antagonists on gallbladder emptying in opossums. Am J Physiol 260:G258–G264PubMed Hanyu N, Dodds WJ, Layman RD, Hogan WJ, Colton DG (1991) Effect of two new cholecystokinin antagonists on gallbladder emptying in opossums. Am J Physiol 260:G258–G264PubMed
43.
go back to reference Miederer SE, Lindstaedt H, Kutz K, Mayershofer R (1979) Efficient treatment of gastric ulcer with proglumide (Milid) in outpatients (double blind trial). Acta Hepatogastroenterol 26:314–318 Miederer SE, Lindstaedt H, Kutz K, Mayershofer R (1979) Efficient treatment of gastric ulcer with proglumide (Milid) in outpatients (double blind trial). Acta Hepatogastroenterol 26:314–318
45.
go back to reference Abu Eid R, Razavi G, Mkrtichyan M, Janik J, Khleif SN (2016) Old-school chemotherapy in immunotherapeutic combination in cancer, a low-cost drug repurposed. Cancer Immunol Res 4:377–382CrossRefPubMed Abu Eid R, Razavi G, Mkrtichyan M, Janik J, Khleif SN (2016) Old-school chemotherapy in immunotherapeutic combination in cancer, a low-cost drug repurposed. Cancer Immunol Res 4:377–382CrossRefPubMed
46.
go back to reference Mkrtichyan M, Najjar YG, Raulfs EC, Abdalla MY, Samara R, Rotem-Yehudar R, Cook L, Khleif SN (2011) Anti-PD-1 synergizes with cyclophosphamide to induce potent anti-tumor vaccine effects through novel mechanisms. Eur J Immunol 41:2977–2986. doi:10.1002/eji.201141639 CrossRefPubMed Mkrtichyan M, Najjar YG, Raulfs EC, Abdalla MY, Samara R, Rotem-Yehudar R, Cook L, Khleif SN (2011) Anti-PD-1 synergizes with cyclophosphamide to induce potent anti-tumor vaccine effects through novel mechanisms. Eur J Immunol 41:2977–2986. doi:10.​1002/​eji.​201141639 CrossRefPubMed
50.
51.
52.
go back to reference Nywening TM, Wang-Gillam A, Sanford DE, Belt BA, Panni RZ, Cusworth BM, Toriola AT, Nieman RK, Worley LA, Yano M, Fowler KJ, Lockhart AC, Suresh R, Tan BR, Lim KH, Fields RC, Strasberg SM, Hawkins WG, Denardo DG, Goedegebuure SP, Linehan DC (2016) Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol 17:651–662. doi:10.1016/S1470-2045(16)00078-4 CrossRefPubMedPubMedCentral Nywening TM, Wang-Gillam A, Sanford DE, Belt BA, Panni RZ, Cusworth BM, Toriola AT, Nieman RK, Worley LA, Yano M, Fowler KJ, Lockhart AC, Suresh R, Tan BR, Lim KH, Fields RC, Strasberg SM, Hawkins WG, Denardo DG, Goedegebuure SP, Linehan DC (2016) Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol 17:651–662. doi:10.​1016/​S1470-2045(16)00078-4 CrossRefPubMedPubMedCentral
53.
go back to reference Hingorani SR, Harris WP, Beck JT, Berdov BA, Wagner SA, Pshevlotsky EM, Tjulandin SA, Gladkov OA, Holcombe RF, Korn R, Raghunand N, Dychter S, Jiang P, Shepard HM, Devoe CE (2016) Phase Ib study of PEGylated recombinant human hyaluronidase and gemcitabine in patients with advanced pancreatic cancer. Clin Cancer Res 22:2848–2854. doi:10.1158/1078-0432.CCR-15-2010 CrossRefPubMed Hingorani SR, Harris WP, Beck JT, Berdov BA, Wagner SA, Pshevlotsky EM, Tjulandin SA, Gladkov OA, Holcombe RF, Korn R, Raghunand N, Dychter S, Jiang P, Shepard HM, Devoe CE (2016) Phase Ib study of PEGylated recombinant human hyaluronidase and gemcitabine in patients with advanced pancreatic cancer. Clin Cancer Res 22:2848–2854. doi:10.​1158/​1078-0432.​CCR-15-2010 CrossRefPubMed
Metadata
Title
Cholecystokinin receptor antagonist alters pancreatic cancer microenvironment and increases efficacy of immune checkpoint antibody therapy in mice
Authors
Jill P. Smith
Shangzi Wang
Sandeep Nadella
Sandra A. Jablonski
Louis M. Weiner
Publication date
01-02-2018
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 2/2018
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-017-2077-9

Other articles of this Issue 2/2018

Cancer Immunology, Immunotherapy 2/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine