Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2020

Open Access 01-12-2020 | Rapid communication

CD13 as a new tumor target for antibody-drug conjugates: validation with the conjugate MI130110

Authors: Juan Manuel Domínguez, Gema Pérez-Chacón, María José Guillén, María José Muñoz-Alonso, Beatriz Somovilla-Crespo, Danay Cibrián, Bárbara Acosta-Iborra, Magdalena Adrados, Cecilia Muñoz-Calleja, Carmen Cuevas, Francisco Sánchez-Madrid, Pablo Avilés, Juan M. Zapata

Published in: Journal of Hematology & Oncology | Issue 1/2020

Login to get access

Abstract

Background

In the search for novel antibody-drug conjugates (ADCs) with therapeutic potential, it is imperative to identify novel targets to direct the antibody moiety. CD13 seems an attractive ADC target as it shows a differential pattern of expression in a variety of tumors and cell lines and it is internalized upon engagement with a suitable monoclonal antibody. PM050489 is a marine cytotoxic compound tightly binding tubulin and impairing microtubule dynamics which is currently undergoing clinical trials for solid tumors.

Methods

Anti-CD13 monoclonal antibody (mAb) TEA1/8 has been used to prepare a novel ADC, MI130110, by conjugation to the marine compound PM050489. In vitro and in vivo experiments have been carried out to demonstrate the activity and specificity of MI130110.

Results

CD13 is readily internalized upon TEA1/8 mAb binding, and the conjugation with PM050489 did not have any effect on the binding or the internalization of the antibody. MI130110 showed remarkable activity and selectivity in vitro on CD13-expressing tumor cells causing the same effects than those described for PM050489, including cell cycle arrest at G2, mitosis with disarrayed and often multipolar spindles consistent with an arrest at metaphase, and induction of cell death. In contrast, none of these toxic effects were observed in CD13-null cell lines incubated with MI130110. Furthermore, in vivo studies showed that MI130110 exhibited excellent antitumor activity in a CD13-positive fibrosarcoma xenograft murine model, with total remissions in a significant number of the treated animals. Mitotic catastrophes, typical of the payload mechanism of action, were also observed in the tumor cells isolated from mice treated with MI130110. In contrast, MI130110 failed to show any activity in a xenograft mouse model of myeloma cells not expressing CD13, thereby corroborating the selectivity of the ADC to its target and its stability in circulation.

Conclusion

Our results show that MI130110 ADC combines the antitumor potential of the PM050489 payload with the selectivity of the TEA1/8 monoclonal anti-CD13 antibody and confirm the correct intracellular processing of the ADC. These results demonstrate the suitability of CD13 as a novel ADC target and the effectiveness of MI130110 as a promising antitumor therapeutic agent.
Appendix
Available only for authorised users
Literature
1.
go back to reference Look AT, Ashmun RA, Shapiro LH, Peiper SC. Human myeloid plasma membrane glycoprotein CD13 (gp150) is identical to aminopeptidase N. J Clin Invest. 1989;83(4):1299–307.PubMedPubMedCentralCrossRef Look AT, Ashmun RA, Shapiro LH, Peiper SC. Human myeloid plasma membrane glycoprotein CD13 (gp150) is identical to aminopeptidase N. J Clin Invest. 1989;83(4):1299–307.PubMedPubMedCentralCrossRef
3.
go back to reference Wickstrom M, Larsson R, Nygren P, Gullbo J. Aminopeptidase N (CD13) as a target for cancer chemotherapy. Cancer Sci. 2011;102(3):501–8.PubMedCrossRef Wickstrom M, Larsson R, Nygren P, Gullbo J. Aminopeptidase N (CD13) as a target for cancer chemotherapy. Cancer Sci. 2011;102(3):501–8.PubMedCrossRef
4.
go back to reference Guzman-Rojas L, Rangel R, Salameh A, Edwards JK, Dondossola E, Kim YG, et al. Cooperative effects of aminopeptidase N (CD13) expressed by nonmalignant and cancer cells within the tumor microenvironment. Proc Natl Acad Sci U S A. 2012;109(5):1637–42.PubMedPubMedCentralCrossRef Guzman-Rojas L, Rangel R, Salameh A, Edwards JK, Dondossola E, Kim YG, et al. Cooperative effects of aminopeptidase N (CD13) expressed by nonmalignant and cancer cells within the tumor microenvironment. Proc Natl Acad Sci U S A. 2012;109(5):1637–42.PubMedPubMedCentralCrossRef
5.
go back to reference Petrovic N, Schacke W, Gahagan JR, O'Conor CA, Winnicka B, Conway RE, et al. CD13/APN regulates endothelial invasion and filopodia formation. Blood. 2007;110(1):142–50.PubMedPubMedCentralCrossRef Petrovic N, Schacke W, Gahagan JR, O'Conor CA, Winnicka B, Conway RE, et al. CD13/APN regulates endothelial invasion and filopodia formation. Blood. 2007;110(1):142–50.PubMedPubMedCentralCrossRef
6.
go back to reference Rangel R, Sun Y, Guzman-Rojas L, Ozawa MG, Sun J, Giordano RJ, et al. Impaired angiogenesis in aminopeptidase N-null mice. Proc Natl Acad Sci U S A. 2007;104(11):4588–93.PubMedPubMedCentralCrossRef Rangel R, Sun Y, Guzman-Rojas L, Ozawa MG, Sun J, Giordano RJ, et al. Impaired angiogenesis in aminopeptidase N-null mice. Proc Natl Acad Sci U S A. 2007;104(11):4588–93.PubMedPubMedCentralCrossRef
7.
go back to reference Di Matteo P, Arrigoni GL, Alberici L, Corti A, Gallo-Stampino C, Traversari C, et al. Enhanced expression of CD13 in vessels of inflammatory and neoplastic tissues. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 2011;59(1):47–59.CrossRef Di Matteo P, Arrigoni GL, Alberici L, Corti A, Gallo-Stampino C, Traversari C, et al. Enhanced expression of CD13 in vessels of inflammatory and neoplastic tissues. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 2011;59(1):47–59.CrossRef
8.
go back to reference Dondossola E, Rangel R, Guzman-Rojas L, Barbu EM, Hosoya H, St John LS, et al. CD13-positive bone marrow-derived myeloid cells promote angiogenesis, tumor growth, and metastasis. Proc Natl Acad Sci U S A. 2013;110(51):20717–22.PubMedPubMedCentralCrossRef Dondossola E, Rangel R, Guzman-Rojas L, Barbu EM, Hosoya H, St John LS, et al. CD13-positive bone marrow-derived myeloid cells promote angiogenesis, tumor growth, and metastasis. Proc Natl Acad Sci U S A. 2013;110(51):20717–22.PubMedPubMedCentralCrossRef
9.
go back to reference Dondossola E, Corti A, Sidman RL, Arap W, Pasqualini R. Bone marrow-derived CD13(+) cells sustain tumor progression: a potential non-malignant target for anticancer therapy. Oncoimmunology. 2014;3:e27716.PubMedPubMedCentralCrossRef Dondossola E, Corti A, Sidman RL, Arap W, Pasqualini R. Bone marrow-derived CD13(+) cells sustain tumor progression: a potential non-malignant target for anticancer therapy. Oncoimmunology. 2014;3:e27716.PubMedPubMedCentralCrossRef
10.
go back to reference Ikeda N, Nakajima Y, Tokuhara T, Hattori N, Sho M, Kanehiro H, et al. Clinical significance of aminopeptidase N/CD13 expression in human pancreatic carcinoma. Clin Cancer Res. 2003;9(4):1503–8.PubMed Ikeda N, Nakajima Y, Tokuhara T, Hattori N, Sho M, Kanehiro H, et al. Clinical significance of aminopeptidase N/CD13 expression in human pancreatic carcinoma. Clin Cancer Res. 2003;9(4):1503–8.PubMed
11.
go back to reference Hashida H, Takabayashi A, Kanai M, Adachi M, Kondo K, Kohno N, et al. Aminopeptidase N is involved in cell motility and angiogenesis: its clinical significance in human colon cancer. Gastroenterology. 2002;122(2):376–86.PubMedCrossRef Hashida H, Takabayashi A, Kanai M, Adachi M, Kondo K, Kohno N, et al. Aminopeptidase N is involved in cell motility and angiogenesis: its clinical significance in human colon cancer. Gastroenterology. 2002;122(2):376–86.PubMedCrossRef
12.
go back to reference Zhang Q, Wang J, Zhang H, Zhao D, Zhang Z, Zhang S. Expression and clinical significance of aminopeptidase N/CD13 in non-small cell lung cancer. J Cancer Res Ther. 2015;11(1):223–8.PubMedCrossRef Zhang Q, Wang J, Zhang H, Zhao D, Zhang Z, Zhang S. Expression and clinical significance of aminopeptidase N/CD13 in non-small cell lung cancer. J Cancer Res Ther. 2015;11(1):223–8.PubMedCrossRef
13.
go back to reference Schmidt LH, Brand C, Stucke-Ring J, Schliemann C, Kessler T, Harrach S, et al. Potential therapeutic impact of CD13 expression in non-small cell lung cancer. PLoS One. 2017;12(6):e0177146.PubMedPubMedCentralCrossRef Schmidt LH, Brand C, Stucke-Ring J, Schliemann C, Kessler T, Harrach S, et al. Potential therapeutic impact of CD13 expression in non-small cell lung cancer. PLoS One. 2017;12(6):e0177146.PubMedPubMedCentralCrossRef
14.
go back to reference Otsuki T, Nakashima T, Hamada H, Takayama Y, Akita S, Masuda T, et al. Aminopeptidase N/CD13 as a potential therapeutic target in malignant pleural mesothelioma. Eur Respir J. 2018;51(5). Otsuki T, Nakashima T, Hamada H, Takayama Y, Akita S, Masuda T, et al. Aminopeptidase N/CD13 as a potential therapeutic target in malignant pleural mesothelioma. Eur Respir J. 2018;51(5).
15.
go back to reference Saida S, Watanabe K, Kato I, Fujino H, Umeda K, Okamoto S, et al. Prognostic significance of aminopeptidase-N (CD13) in hepatoblastoma. Pediatr Int. 2015;57(4):558–66.PubMedCrossRef Saida S, Watanabe K, Kato I, Fujino H, Umeda K, Okamoto S, et al. Prognostic significance of aminopeptidase-N (CD13) in hepatoblastoma. Pediatr Int. 2015;57(4):558–66.PubMedCrossRef
16.
go back to reference Kessler T, Baumeier A, Brand C, Grau M, Angenendt L, Harrach S, et al. Aminopeptidase N (CD13): Expression, Prognostic Impact, and Use as Therapeutic Target for Tissue Factor Induced Tumor Vascular Infarction in Soft Tissue Sarcoma. Transl Oncol. 2018;11(6):1271–82.PubMedPubMedCentralCrossRef Kessler T, Baumeier A, Brand C, Grau M, Angenendt L, Harrach S, et al. Aminopeptidase N (CD13): Expression, Prognostic Impact, and Use as Therapeutic Target for Tissue Factor Induced Tumor Vascular Infarction in Soft Tissue Sarcoma. Transl Oncol. 2018;11(6):1271–82.PubMedPubMedCentralCrossRef
17.
go back to reference Piedfer M, Dauzonne D, Tang R, N'Guyen J, Billard C, Bauvois B. Aminopeptidase-N/CD13 is a potential proapoptotic target in human myeloid tumor cells. FASEB J. 2011;25(8):2831–42.PubMedCrossRef Piedfer M, Dauzonne D, Tang R, N'Guyen J, Billard C, Bauvois B. Aminopeptidase-N/CD13 is a potential proapoptotic target in human myeloid tumor cells. FASEB J. 2011;25(8):2831–42.PubMedCrossRef
18.
go back to reference Yu B, Liu D. Antibody-drug conjugates in clinical trials for lymphoid malignancies and multiple myeloma. Journal of hematology & oncology. 2019;12(1):94.CrossRef Yu B, Liu D. Antibody-drug conjugates in clinical trials for lymphoid malignancies and multiple myeloma. Journal of hematology & oncology. 2019;12(1):94.CrossRef
19.
go back to reference Abdollahpour-Alitappeh M, Lotfinia M, Gharibi T, Mardaneh J, Farhadihosseinabadi B, Larki P, et al. Antibody-drug conjugates (ADCs) for cancer therapy: Strategies, challenges, and successes. J Cell Physiol. 2019;234(5):5628–42.PubMedCrossRef Abdollahpour-Alitappeh M, Lotfinia M, Gharibi T, Mardaneh J, Farhadihosseinabadi B, Larki P, et al. Antibody-drug conjugates (ADCs) for cancer therapy: Strategies, challenges, and successes. J Cell Physiol. 2019;234(5):5628–42.PubMedCrossRef
20.
go back to reference Wolska-Washer A. Robak T. Drug Saf: Safety and Tolerability of Antibody-Drug Conjugates in Cancer; 2019. Wolska-Washer A. Robak T. Drug Saf: Safety and Tolerability of Antibody-Drug Conjugates in Cancer; 2019.
21.
go back to reference Beck A, Goetsch L, Dumontet C, Corvaia N. Strategies and challenges for the next generation of antibody-drug conjugates. Nat Rev Drug Discov. 2017;16(5):315–37.PubMedCrossRef Beck A, Goetsch L, Dumontet C, Corvaia N. Strategies and challenges for the next generation of antibody-drug conjugates. Nat Rev Drug Discov. 2017;16(5):315–37.PubMedCrossRef
22.
go back to reference Lambert JM, Berkenblit A. Antibody-Drug Conjugates for Cancer Treatment. Annu Rev Med. 2018;69:191–207.PubMedCrossRef Lambert JM, Berkenblit A. Antibody-Drug Conjugates for Cancer Treatment. Annu Rev Med. 2018;69:191–207.PubMedCrossRef
23.
go back to reference Bouchet S, Tang R, Fava F, Legrand O, Bauvois B. Targeting CD13 (aminopeptidase-N) in turn downregulates ADAM17 by internalization in acute myeloid leukaemia cells. Oncotarget. 2014;5(18):8211–22.PubMedPubMedCentralCrossRef Bouchet S, Tang R, Fava F, Legrand O, Bauvois B. Targeting CD13 (aminopeptidase-N) in turn downregulates ADAM17 by internalization in acute myeloid leukaemia cells. Oncotarget. 2014;5(18):8211–22.PubMedPubMedCentralCrossRef
24.
go back to reference Miki T, Takegami Y, Okawa K, Muraguchi T, Noda M, Takahashi C. The reversion-inducing cysteine-rich protein with Kazal motifs (RECK) interacts with membrane type 1 matrix metalloproteinase and CD13/aminopeptidase N and modulates their endocytic pathways. J Biol Chem. 2007;282(16):12341–52.PubMedCrossRef Miki T, Takegami Y, Okawa K, Muraguchi T, Noda M, Takahashi C. The reversion-inducing cysteine-rich protein with Kazal motifs (RECK) interacts with membrane type 1 matrix metalloproteinase and CD13/aminopeptidase N and modulates their endocytic pathways. J Biol Chem. 2007;282(16):12341–52.PubMedCrossRef
25.
go back to reference Martin MJ, Coello L, Fernandez R, Reyes F, Rodriguez A, Murcia C, et al. Isolation and first total synthesis of PM050489 and PM060184, two new marine anticancer compounds. J Am Chem Soc. 2013;135(27):10164–71.PubMedCrossRef Martin MJ, Coello L, Fernandez R, Reyes F, Rodriguez A, Murcia C, et al. Isolation and first total synthesis of PM050489 and PM060184, two new marine anticancer compounds. J Am Chem Soc. 2013;135(27):10164–71.PubMedCrossRef
26.
go back to reference Prota AE, Bargsten K, Diaz JF, Marsh M, Cuevas C, Liniger M, et al. A new tubulin-binding site and pharmacophore for microtubule-destabilizing anticancer drugs. Proc Natl Acad Sci U S A. 2014;111(38):13817–21.PubMedPubMedCentralCrossRef Prota AE, Bargsten K, Diaz JF, Marsh M, Cuevas C, Liniger M, et al. A new tubulin-binding site and pharmacophore for microtubule-destabilizing anticancer drugs. Proc Natl Acad Sci U S A. 2014;111(38):13817–21.PubMedPubMedCentralCrossRef
27.
go back to reference Aviles P, Dominguez JM, Guillen MJ, Munoz-Alonso MJ, Mateo C, Rodriguez-Acebes R, et al. MI130004, a novel antibody-drug conjugate combining trastuzumab with a molecule of marine origin, shows outstanding in vivo activity against HER2-expressing tumors. Mol Cancer Ther. 2018;17(4):786–94.PubMedCrossRef Aviles P, Dominguez JM, Guillen MJ, Munoz-Alonso MJ, Mateo C, Rodriguez-Acebes R, et al. MI130004, a novel antibody-drug conjugate combining trastuzumab with a molecule of marine origin, shows outstanding in vivo activity against HER2-expressing tumors. Mol Cancer Ther. 2018;17(4):786–94.PubMedCrossRef
28.
go back to reference Martin N, Ma D, Herbet A, Boquet D, Winnik FM, Tribet C. Prevention of thermally induced aggregation of IgG antibodies by noncovalent interaction with poly(acrylate) derivatives. Biomacromolecules. 2014;15(8):2952–62.PubMedCrossRef Martin N, Ma D, Herbet A, Boquet D, Winnik FM, Tribet C. Prevention of thermally induced aggregation of IgG antibodies by noncovalent interaction with poly(acrylate) derivatives. Biomacromolecules. 2014;15(8):2952–62.PubMedCrossRef
29.
go back to reference Springer TA, Kitayama J. Endothelial cell antigens: Section report. In: Knapp W, Dörken B, Gilks WR, Rieber EP, Schmidt RE, Stein H, et al., editors. Leukocyte typing IV: white cells differentiation antigens. New York: Oxfod University Press; 1990. Springer TA, Kitayama J. Endothelial cell antigens: Section report. In: Knapp W, Dörken B, Gilks WR, Rieber EP, Schmidt RE, Stein H, et al., editors. Leukocyte typing IV: white cells differentiation antigens. New York: Oxfod University Press; 1990.
30.
go back to reference Perez-Chacon G, de Los RC, Zapata JM. Indole-3-carbinol induces cMYC and IAP-family downmodulation and promotes apoptosis of Epstein-Barr virus (EBV)-positive but not of EBV-negative Burkitt’s lymphoma cell lines. Pharmacological research : the official journal of the Italian Pharmacological Society. 2014;89:46–56.CrossRef Perez-Chacon G, de Los RC, Zapata JM. Indole-3-carbinol induces cMYC and IAP-family downmodulation and promotes apoptosis of Epstein-Barr virus (EBV)-positive but not of EBV-negative Burkitt’s lymphoma cell lines. Pharmacological research : the official journal of the Italian Pharmacological Society. 2014;89:46–56.CrossRef
31.
go back to reference Martinez-Diez M, Guillen-Navarro MJ, Pera B, Bouchet BP, Martinez-Leal JF, Barasoain I, et al. PM060184, a new tubulin binding agent with potent antitumor activity including P-glycoprotein over-expressing tumors. Biochem Pharmacol. 2014;88(3):291–302.PubMedCrossRef Martinez-Diez M, Guillen-Navarro MJ, Pera B, Bouchet BP, Martinez-Leal JF, Barasoain I, et al. PM060184, a new tubulin binding agent with potent antitumor activity including P-glycoprotein over-expressing tumors. Biochem Pharmacol. 2014;88(3):291–302.PubMedCrossRef
32.
go back to reference Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, et al. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009;16(1):3–11.PubMedCrossRef Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, et al. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009;16(1):3–11.PubMedCrossRef
33.
go back to reference Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Proteomics. Tissue-based map of the human proteome. Science. 2015;347(6220):1260419. Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Proteomics. Tissue-based map of the human proteome. Science. 2015;347(6220):1260419.
34.
go back to reference Uhlen M, Zhang C, Lee S, Sjostedt E, Fagerberg L, Bidkhori G, et al. A pathology atlas of the human cancer transcriptome. Science. 2017;357(6352). Uhlen M, Zhang C, Lee S, Sjostedt E, Fagerberg L, Bidkhori G, et al. A pathology atlas of the human cancer transcriptome. Science. 2017;357(6352).
35.
go back to reference Yamanaka C, Wada H, Eguchi H, Hatano H, Gotoh K, Noda T, et al. Clinical significance of CD13 and epithelial mesenchymal transition (EMT) markers in hepatocellular carcinoma. Jpn J Clin Oncol. 2018;48(1):52–60.PubMedCrossRef Yamanaka C, Wada H, Eguchi H, Hatano H, Gotoh K, Noda T, et al. Clinical significance of CD13 and epithelial mesenchymal transition (EMT) markers in hepatocellular carcinoma. Jpn J Clin Oncol. 2018;48(1):52–60.PubMedCrossRef
36.
go back to reference Brunelli M, Erdini F, Cima L, Eccher A, Fioravanzo A, Gobbo S, et al. Proximal CD13 versus distal GATA-3 Expression in renal neoplasia according to WHO 2016 classification. Appl Immunohistochem Mol Morphol. 2018;26(5):316–23.PubMedCrossRef Brunelli M, Erdini F, Cima L, Eccher A, Fioravanzo A, Gobbo S, et al. Proximal CD13 versus distal GATA-3 Expression in renal neoplasia according to WHO 2016 classification. Appl Immunohistochem Mol Morphol. 2018;26(5):316–23.PubMedCrossRef
37.
go back to reference Nohara S, Kato K, Fujiwara D, Sakuragi N, Yanagihara K, Iwanuma Y, et al. Aminopeptidase N (APN/CD13) as a target molecule for scirrhous gastric cancer. Clin Res Hepatol Gastroenterol. 2016;40(4):494–503.PubMedCrossRefPubMedCentral Nohara S, Kato K, Fujiwara D, Sakuragi N, Yanagihara K, Iwanuma Y, et al. Aminopeptidase N (APN/CD13) as a target molecule for scirrhous gastric cancer. Clin Res Hepatol Gastroenterol. 2016;40(4):494–503.PubMedCrossRefPubMedCentral
38.
go back to reference Raimbault A, Machherndl-Spandl S, Itzykson R, Clauser S, Chapuis N, Mathis S, et al. CD13 expression in B cell malignancies is a hallmark of plasmacytic differentiation. Br J Haematol. 2019;184(4):625–33.PubMedCrossRef Raimbault A, Machherndl-Spandl S, Itzykson R, Clauser S, Chapuis N, Mathis S, et al. CD13 expression in B cell malignancies is a hallmark of plasmacytic differentiation. Br J Haematol. 2019;184(4):625–33.PubMedCrossRef
39.
go back to reference Williams BA, Law A, Hunyadkurti J, Desilets S, Leyton JV, Keating A. Antibody therapies for acute myeloid leukemia: unconjugated, toxin-conjugated, radio-conjugated and multivalent formats. J Clin Med. 2019;8(8). Williams BA, Law A, Hunyadkurti J, Desilets S, Leyton JV, Keating A. Antibody therapies for acute myeloid leukemia: unconjugated, toxin-conjugated, radio-conjugated and multivalent formats. J Clin Med. 2019;8(8).
40.
go back to reference Maynadie M, De Angelis R, Marcos-Gragera R, Visser O, Allemani C, Tereanu C, et al. Survival of European patients diagnosed with myeloid malignancies: a HAEMACARE study. Haematologica. 2013;98(2):230–8.PubMedPubMedCentralCrossRef Maynadie M, De Angelis R, Marcos-Gragera R, Visser O, Allemani C, Tereanu C, et al. Survival of European patients diagnosed with myeloid malignancies: a HAEMACARE study. Haematologica. 2013;98(2):230–8.PubMedPubMedCentralCrossRef
41.
go back to reference Huyghe L, Van Parys A, Cauwels A, Van Lint S, De Munter S, Bultinck J, et al. Safe eradication of large established tumors using neovasculature-targeted tumor necrosis factor-based therapies. EMBO Mol Med. 2020:e11223. Huyghe L, Van Parys A, Cauwels A, Van Lint S, De Munter S, Bultinck J, et al. Safe eradication of large established tumors using neovasculature-targeted tumor necrosis factor-based therapies. EMBO Mol Med. 2020:e11223.
42.
go back to reference Pasqualini R, Koivunen E, Kain R, Lahdenranta J, Sakamoto M, Stryhn A, et al. Aminopeptidase N is a receptor for tumor-homing peptides and a target for inhibiting angiogenesis. Cancer Res. 2000;60(3):722–7.PubMedPubMedCentral Pasqualini R, Koivunen E, Kain R, Lahdenranta J, Sakamoto M, Stryhn A, et al. Aminopeptidase N is a receptor for tumor-homing peptides and a target for inhibiting angiogenesis. Cancer Res. 2000;60(3):722–7.PubMedPubMedCentral
43.
go back to reference van Hensbergen Y, Broxterman HJ, Elderkamp YW, Lankelma J, Beers JC, Heijn M, et al. A doxorubicin-CNGRC-peptide conjugate with prodrug properties. Biochemical pharmacology. 2002;63(5):897–908.PubMedCrossRef van Hensbergen Y, Broxterman HJ, Elderkamp YW, Lankelma J, Beers JC, Heijn M, et al. A doxorubicin-CNGRC-peptide conjugate with prodrug properties. Biochemical pharmacology. 2002;63(5):897–908.PubMedCrossRef
44.
go back to reference Mukhopadhyay S, Barnes CM, Haskel A, Short SM, Barnes KR, Lippard SJ. Conjugated platinum(IV)-peptide complexes for targeting angiogenic tumor vasculature. Bioconjug Chem. 2008;19(1):39–49.PubMedCrossRef Mukhopadhyay S, Barnes CM, Haskel A, Short SM, Barnes KR, Lippard SJ. Conjugated platinum(IV)-peptide complexes for targeting angiogenic tumor vasculature. Bioconjug Chem. 2008;19(1):39–49.PubMedCrossRef
45.
go back to reference Zheng YB, Gong JH, Liu XJ, Li Y, Zhen YS. A CD13-targeting peptide integrated protein inhibits human liver cancer growth by killing cancer stem cells and suppressing angiogenesis. Mol Carcinog. 2017;56(5):1395–404.PubMedCrossRef Zheng YB, Gong JH, Liu XJ, Li Y, Zhen YS. A CD13-targeting peptide integrated protein inhibits human liver cancer growth by killing cancer stem cells and suppressing angiogenesis. Mol Carcinog. 2017;56(5):1395–404.PubMedCrossRef
46.
go back to reference Gregorc V, Santoro A, Bennicelli E, Punt CJ, Citterio G, Timmer-Bonte JN, et al. Phase Ib study of NGR-hTNF, a selective vascular targeting agent, administered at low doses in combination with doxorubicin to patients with advanced solid tumours. Br J Cancer. 2009;101(2):219–24.PubMedPubMedCentralCrossRef Gregorc V, Santoro A, Bennicelli E, Punt CJ, Citterio G, Timmer-Bonte JN, et al. Phase Ib study of NGR-hTNF, a selective vascular targeting agent, administered at low doses in combination with doxorubicin to patients with advanced solid tumours. Br J Cancer. 2009;101(2):219–24.PubMedPubMedCentralCrossRef
47.
go back to reference Bieker R, Kessler T, Schwoppe C, Padro T, Persigehl T, Bremer C, et al. Infarction of tumor vessels by NGR-peptide-directed targeting of tissue factor: experimental results and first-in-man experience. Blood. 2009;113(20):5019–27.PubMedCrossRef Bieker R, Kessler T, Schwoppe C, Padro T, Persigehl T, Bremer C, et al. Infarction of tumor vessels by NGR-peptide-directed targeting of tissue factor: experimental results and first-in-man experience. Blood. 2009;113(20):5019–27.PubMedCrossRef
48.
go back to reference Grieger E, Gresch G, Niesen J, Woitok M, Barth S, Fischer R, et al. Efficient targeting of CD13 on cancer cells by the immunotoxin scFv13-ETA' and the bispecific scFv [13xds16]. J Cancer Res Clin Oncol. 2017;143(11):2159–70.PubMedCrossRef Grieger E, Gresch G, Niesen J, Woitok M, Barth S, Fischer R, et al. Efficient targeting of CD13 on cancer cells by the immunotoxin scFv13-ETA' and the bispecific scFv [13xds16]. J Cancer Res Clin Oncol. 2017;143(11):2159–70.PubMedCrossRef
49.
go back to reference Austin CD, De Maziere AM, Pisacane PI, van Dijk SM, Eigenbrot C, Sliwkowski MX, et al. Endocytosis and sorting of ErbB2 and the site of action of cancer therapeutics trastuzumab and geldanamycin. Mol Biol Cell. 2004;15(12):5268–82.PubMedPubMedCentralCrossRef Austin CD, De Maziere AM, Pisacane PI, van Dijk SM, Eigenbrot C, Sliwkowski MX, et al. Endocytosis and sorting of ErbB2 and the site of action of cancer therapeutics trastuzumab and geldanamycin. Mol Biol Cell. 2004;15(12):5268–82.PubMedPubMedCentralCrossRef
50.
go back to reference Hommelgaard AM, Lerdrup M, van Deurs B. Association with membrane protrusions makes ErbB2 an internalization-resistant receptor. Mol Biol Cell. 2004;15(4):1557–67.PubMedPubMedCentralCrossRef Hommelgaard AM, Lerdrup M, van Deurs B. Association with membrane protrusions makes ErbB2 an internalization-resistant receptor. Mol Biol Cell. 2004;15(4):1557–67.PubMedPubMedCentralCrossRef
51.
go back to reference Maass KF, Kulkarni C, Betts AM, Wittrup KD. Determination of cellular processing rates for a trastuzumab-maytansinoid antibody-drug conjugate (ADC) highlights key parameters for ADC design. Aaps j. 2016;18(3):635–46.PubMedPubMedCentralCrossRef Maass KF, Kulkarni C, Betts AM, Wittrup KD. Determination of cellular processing rates for a trastuzumab-maytansinoid antibody-drug conjugate (ADC) highlights key parameters for ADC design. Aaps j. 2016;18(3):635–46.PubMedPubMedCentralCrossRef
52.
go back to reference Okeley NM, Miyamoto JB, Zhang X, Sanderson RJ, Benjamin DR, Sievers EL, et al. Intracellular activation of SGN-35, a potent anti-CD30 antibody-drug conjugate. Clin Cancer Res. 2010;16(3):888–97.PubMedCrossRef Okeley NM, Miyamoto JB, Zhang X, Sanderson RJ, Benjamin DR, Sievers EL, et al. Intracellular activation of SGN-35, a potent anti-CD30 antibody-drug conjugate. Clin Cancer Res. 2010;16(3):888–97.PubMedCrossRef
53.
go back to reference Jager E, van der Velden VH, te Marvelde JG, Walter RB, Agur Z, Vainstein V. Targeted drug delivery by gemtuzumab ozogamicin: mechanism-based mathematical model for treatment strategy improvement and therapy individualization. PLoS One. 2011;6(9):e24265.PubMedPubMedCentralCrossRef Jager E, van der Velden VH, te Marvelde JG, Walter RB, Agur Z, Vainstein V. Targeted drug delivery by gemtuzumab ozogamicin: mechanism-based mathematical model for treatment strategy improvement and therapy individualization. PLoS One. 2011;6(9):e24265.PubMedPubMedCentralCrossRef
54.
go back to reference van Der Velden VH, te Marvelde JG, Hoogeveen PG, Bernstein ID, Houtsmuller AB, Berger MS, et al. Targeting of the CD33-calicheamicin immunoconjugate Mylotarg (CMA-676) in acute myeloid leukemia: in vivo and in vitro saturation and internalization by leukemic and normal myeloid cells. Blood. 2001;97(10):3197–204.CrossRef van Der Velden VH, te Marvelde JG, Hoogeveen PG, Bernstein ID, Houtsmuller AB, Berger MS, et al. Targeting of the CD33-calicheamicin immunoconjugate Mylotarg (CMA-676) in acute myeloid leukemia: in vivo and in vitro saturation and internalization by leukemic and normal myeloid cells. Blood. 2001;97(10):3197–204.CrossRef
55.
go back to reference Durbin KR, Phipps C, Liao X. Mechanistic modeling of antibody-drug conjugate internalization at the cellular level reveals inefficient processing steps. Mol Cancer Ther. 2018;17(6):1341–51.PubMedCrossRef Durbin KR, Phipps C, Liao X. Mechanistic modeling of antibody-drug conjugate internalization at the cellular level reveals inefficient processing steps. Mol Cancer Ther. 2018;17(6):1341–51.PubMedCrossRef
56.
go back to reference Ackerman ME, Pawlowski D, Wittrup KD. Effect of antigen turnover rate and expression level on antibody penetration into tumor spheroids. Mol Cancer Ther. 2008;7(7):2233–40.PubMedPubMedCentralCrossRef Ackerman ME, Pawlowski D, Wittrup KD. Effect of antigen turnover rate and expression level on antibody penetration into tumor spheroids. Mol Cancer Ther. 2008;7(7):2233–40.PubMedPubMedCentralCrossRef
57.
go back to reference van der Jagt RH, Badger CC, Appelbaum FR, Press OW, Matthews DC, Eary JF, et al. Localization of radiolabeled antimyeloid antibodies in a human acute leukemia xenograft tumor model. Cancer Res. 1992;52(1):89–94.PubMed van der Jagt RH, Badger CC, Appelbaum FR, Press OW, Matthews DC, Eary JF, et al. Localization of radiolabeled antimyeloid antibodies in a human acute leukemia xenograft tumor model. Cancer Res. 1992;52(1):89–94.PubMed
58.
go back to reference Bostrom J, Haber L, Koenig P, Kelley RF, Fuh G. High affinity antigen recognition of the dual specific variants of herceptin is entropy-driven in spite of structural plasticity. PLoS One. 2011;6(4):e17887.PubMedPubMedCentralCrossRef Bostrom J, Haber L, Koenig P, Kelley RF, Fuh G. High affinity antigen recognition of the dual specific variants of herceptin is entropy-driven in spite of structural plasticity. PLoS One. 2011;6(4):e17887.PubMedPubMedCentralCrossRef
59.
go back to reference Elmlund L, Kack C, Aastrup T, Nicholls IA. Study of the interaction of trastuzumab and SKOV3 epithelial cancer cells using a quartz crystal microbalance sensor. Sensors (Basel). 2015;15(3):5884–94.PubMedPubMedCentralCrossRef Elmlund L, Kack C, Aastrup T, Nicholls IA. Study of the interaction of trastuzumab and SKOV3 epithelial cancer cells using a quartz crystal microbalance sensor. Sensors (Basel). 2015;15(3):5884–94.PubMedPubMedCentralCrossRef
60.
go back to reference Sun MM, Beam KS, Cerveny CG, Hamblett KJ, Blackmore RS, Torgov MY, et al. Reduction-alkylation strategies for the modification of specific monoclonal antibody disulfides. Bioconjug Chem. 2005;16(5):1282–90.PubMedPubMedCentralCrossRef Sun MM, Beam KS, Cerveny CG, Hamblett KJ, Blackmore RS, Torgov MY, et al. Reduction-alkylation strategies for the modification of specific monoclonal antibody disulfides. Bioconjug Chem. 2005;16(5):1282–90.PubMedPubMedCentralCrossRef
61.
go back to reference Ritchie M, Tchistiakova L, Scott N. Implications of receptor-mediated endocytosis and intracellular trafficking dynamics in the development of antibody drug conjugates. MAbs. 2013;5(1):13–21.PubMedPubMedCentralCrossRef Ritchie M, Tchistiakova L, Scott N. Implications of receptor-mediated endocytosis and intracellular trafficking dynamics in the development of antibody drug conjugates. MAbs. 2013;5(1):13–21.PubMedPubMedCentralCrossRef
62.
go back to reference Schier R, McCall A, Adams GP, Marshall KW, Merritt H, Yim M, et al. Isolation of picomolar affinity anti-c-erbB-2 single-chain Fv by molecular evolution of the complementarity determining regions in the center of the antibody binding site. J Mol Biol. 1996;263(4):551–67.PubMedCrossRef Schier R, McCall A, Adams GP, Marshall KW, Merritt H, Yim M, et al. Isolation of picomolar affinity anti-c-erbB-2 single-chain Fv by molecular evolution of the complementarity determining regions in the center of the antibody binding site. J Mol Biol. 1996;263(4):551–67.PubMedCrossRef
63.
go back to reference Rudnick SI, Lou J, Shaller CC, Tang Y, Klein-Szanto AJ, Weiner LM, et al. Influence of affinity and antigen internalization on the uptake and penetration of Anti-HER2 antibodies in solid tumors. Cancer Res. 2011;71(6):2250–9.PubMedPubMedCentralCrossRef Rudnick SI, Lou J, Shaller CC, Tang Y, Klein-Szanto AJ, Weiner LM, et al. Influence of affinity and antigen internalization on the uptake and penetration of Anti-HER2 antibodies in solid tumors. Cancer Res. 2011;71(6):2250–9.PubMedPubMedCentralCrossRef
65.
Metadata
Title
CD13 as a new tumor target for antibody-drug conjugates: validation with the conjugate MI130110
Authors
Juan Manuel Domínguez
Gema Pérez-Chacón
María José Guillén
María José Muñoz-Alonso
Beatriz Somovilla-Crespo
Danay Cibrián
Bárbara Acosta-Iborra
Magdalena Adrados
Cecilia Muñoz-Calleja
Carmen Cuevas
Francisco Sánchez-Madrid
Pablo Avilés
Juan M. Zapata
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2020
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-00865-7

Other articles of this Issue 1/2020

Journal of Hematology & Oncology 1/2020 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