Trop-2 is a novel target for solid cancer therapy with sacituzumab govitecan (IMMU-132), an antibody-drug conjugate (ADC)

David M Goldenberg, Thomas M Cardillo, Serengulam V Govindan, Edmund A Rossi, Robert M Sharkey, David M Goldenberg, Thomas M Cardillo, Serengulam V Govindan, Edmund A Rossi, Robert M Sharkey

Abstract

Trop-2 is a novel target for ADC therapy because of its high expression by many solid cancers. The rational development of IMMU-132 represents a paradigm shift as an ADC that binds a well-known moderately-cytotoxic drug, SN-38, to the anti-Trop-2 antibody. In vitro and in vivo studies show enhanced efficacy, while there is a gradual release of SN-38 that contributes to the overall effect. IMMU-132 is most efficacious at a high drug:antibody ratio (DAR) of 7.6:1, which does not affect binding and pharmacokinetics. It targets up to 136-fold more SN-38 to a human cancer xenograft than irinotecan, SN-38's prodrug. IMMU-132 delivers SN-38 in its most active, non-glucuronidated form, which may explain the lower frequency of severe diarrhea than with irinotecan. Thus, this ADC, carrying a moderately-toxic drug targeting Trop-2 represents a novel cancer therapeutic that is showing promising activity in patients with several metastatic cancer types, including triple-negative breast cancer, non-small-cell and small-cell lung cancers.

Keywords: SN-38; Trop-2; antibody-drug conjugate; solid cancers; triple-negative breast cancer.

Conflict of interest statement

CONFLICTS OF INTEREST

All authors have employment and stock or stock options in Immunomedics, Inc.

Figures

Figure 1. Structures of irinotecan, SN-38, and…
Figure 1. Structures of irinotecan, SN-38, and CL2A-SN-38
A. Irinotecan schematic indicates the molecule's 7, 10, & 20 positions. B. IMMU-132 ADC shows SN-38 positioning, site of SN-38 cleavage, PEG moiety to enhance solubility, and site coupled to the antibody. C.In vitro serum stability of IMMU-132 in monkey or human serum.
Figure 2. Hydrophobic interaction chromatography of IMMU-132
Figure 2. Hydrophobic interaction chromatography of IMMU-132
Representative HIC-HPLC trace of IMMU-132 resolved with a 15-min gradient of 2.25 M to 1.5 M NaCl. Peaks representing species with DARs of 6, 7 and 8 are indicated.
Figure 3. Therapeutic efficacy of IMMU-132 with…
Figure 3. Therapeutic efficacy of IMMU-132 with different DARs
NCI-N87 human gastric carcinoma xenografts (subcutaneous) were established as described in Materials and Methods. A. Four groups of mice (N = 9) were injected IV with 2 × 0.5 mg (arrows) of IMMU-132 conjugates prepared with a DAR = 6.89, 3.28, or 1.64. Control animals received saline. Therapy began 7 days after tumor cells were administered (size was 0.248 ± 0.047cm3). Survival curves were generated based on the time to progression to ≥ 1.0 cm3, and were analyzed by log-rank test (significance at P ≤ 0.05). B. NCI-N87 tumor-bearing mice (N = 7–9; starting size = 0.271 ± 0.053cm3) were treated with either 0.5 mg IMMU-132 (DAR = 6.89) or 1.0 mg DAR = 3.28 twice weekly for two weeks (arrows). Mice were euthanized and deemed to have succumbed to disease once tumors grew to > 1.0 cm3. Profiles of individual tumor growth were obtained through linear-curve modeling. Statistical analysis of tumor growth was based on area under the curve (AUC) performed up to the time that the first animal within a group was euthanized due to disease progression. An f-test was employed to determine equality of variance between groups prior to statistical analysis of growth curves. A two-tailed t-test was used to assess statistical significance between the various treatment groups and controls, except for the saline control, where a one-tailed t-test was used (significance at P ≤ 0.05).
Figure 4. Immunohistology of TNBC patient specimens
Figure 4. Immunohistology of TNBC patient specimens
Strong (3+) Trop-2 expression in two TNBC specimens within a tumor microarray.
Figure 5. Therapeutic efficacy of IMMU-132 in…
Figure 5. Therapeutic efficacy of IMMU-132 in TNBC xenograft models
A. Twenty-two days after subcutaneously implantation of MDA-MB-468 tumors (at the onset of treatment, tumors averaged 0.223 ± 0.055 cm3), nude mice (7–8 per group) were injected with IMMU-132 or a control hA20 anti-CD20-SN-38 conjugate twice weekly for two weeks (0.12 or 0.20 mg/kg SN-38 equivalents per dose). Other animals were given irinotecan (10 mg/kg/dose; SN-38 equivalent based on mass = 5.8 mg/kg) every other day for 10 days for a total of five injections. Statistical analysis performed in the same manner as mentioned in Fig 2B. B. Starting on day 56 after treatment initiation, all animals in the control hA20-SN-38 group were given IMMU-132 (4 × 0.2 mg/kg SN-38 equivalents). The size of the tumors in the individual animals of this group from the onset of tumor transplantation is given. Purple arrows indicate when the hA20-SN-38 conjugate was first given, and red arrows indicate when the treatment with IMMU-132 was initiated. C. Mice (N = 12) bearing the MDA-MB-231 TNBC cell line (0.335 ± 0.078 cm3) were treated with IMMU-132 or the control hA20-SN-38 conjugate (0.4 mg/kg SN-38 equivalents), irinotecan (6.5 mg/kg; ∼3.8 mg/kg SN-38 equivalents), or a combination of hRS7 IgG (25 mg/kg) plus irinotecan (6.5 mg/kg).
Figure 6. IMMU-132 mediated pro-apoptosis signaling in…
Figure 6. IMMU-132 mediated pro-apoptosis signaling in human breast cancer lines
MDA-MB-468 or SK-BR-3 cells were exposed to 1 μM SN-38, the SN-38-equivalent of IMMU-132, or protein equivalent of hRS7 for the indicated times. Cells were harvested and Western blots performed as described in Materials and Methods. Untreated control cells (control) were maintained in growth medium alone until harvested after 48 h. Blots shown are representative of one of two separate experiments.
Figure 7. Determination of SN-38 and associated…
Figure 7. Determination of SN-38 and associated products in serum and Capan-1 tumors taken from animals given irinotecan or IMMU-132
Animals were given irinotecan (773 μg) or IMMU-132 (1.0 mg) and then at 5 intervals, 3 animals from each group were euthanized with serum panel (A) and tumor panel (B) extracted for the products of interest. Data are only shown for sampling intervals where detectable product was measured (e.g., animals given irinotecan did not have any detectable products at the 24-h sampling interval).

References

    1. Basu A, Goldenberg DM, Stein R. The epithelial/carcinoma antigen EGP-1, recognized by monoclonal antibody RS7–3G11, is phosphorylated on serine 303. Int J Cancer. 1995;62:472–479.
    1. Cardillo TM, Govindan SV, Sharkey RM, Trisal P, Goldenberg DM. Humanized anti-Trop-2 IgG-SN-38 conjugate for effective treatment of diverse epithelial cancers: preclinical studies in human cancer xenograft models and monkeys. Clin Cancer Res. 2011;17:3157–3169.
    1. Stein R, Chen S, Sharkey RM, Goldenberg DM. Murine monoclonal antibodies raised against human non-small cell carcinoma of the lung: specificity and tumor targeting. Cancer Res. 1990;50:1330–1336.
    1. Stein R, Basu A, Chen S, Shih LB, Goldenberg DM. Specificity and properties of MAb RS7–3G11 and the antigen defined by this pancarcinoma monoclonal antibody. Int J Cancer. 1993;55:938–946.
    1. Calabrese G, Crescenzi C, Morizio E, Palka G, Guerra E, Alberti S. Assignment of TACSTD1 (alias TROP1, M4S1) to human chromosome 2p21 and refinement of mapping of TACSTD2 (alias TROP2, M1S1) to human chromosome 1p32 by in situ hybridization. Cytogenet Cell Genet. 2001;92:164–165.
    1. Linnenbach AJ, Seng BA, Wu S, Robbins S, Scollon M, Pyrc JJ, Druck T, Huebner K. Retroposition in a family of carcinoma-associated antigen genes. Mol Cell Biol. 1993;13:1507–1515.
    1. Stein R, Basu A, Goldenberg DM, Lloyd KO, Mattes MJ. Characterization of cluster 13: the epithelial/carcinoma antigen recognized by MAb RS7. Int J Cancer Suppl. 1994;8:98–102.
    1. Fornaro M, Dell'Arciprete R, Stella M, Bucci C, Nutini M, Capri MG, Alberti S. Cloning of the gene encoding Trop-2, a cell-surface glycoprotein expressed by human carcinomas. Int J Cancer. 1995;62:610–618.
    1. Lipinski M, Parks DR, Rouse RV, Herzenberg LA. Human trophoblast cell-surface antigens defined by monoclonal antibodies. Proc Natl Acad Sci USA. 1981;78:5147–5150.
    1. Miotti S, Canevari S, Menard S, Mezzanzanica D, Porro G, Pupa SM, Regazzoni M, Tagliabue E, Colnaghi MI. Characterization of human ovarian carcinoma-associated antigens defined by novel monoclonal antibodies with tumor-restricted specificity. Int J Cancer. 1987;39:297–303.
    1. Ripani E, Sacchetti A, Corda D, Alberti S. Human Trop-2 is a tumor-associated calcium signal transducer. Int J Cancer. 1998;76:671–676.
    1. Cubas R, Li M, Chen C, Yao Q. Trop2: a possible therapeutic target for late stage epithelial carcinomas. Biochim Biophys Acta. 2009;1796:309–314.
    1. Trerotola M, Cantanelli P, Guerra E, Tripaldi R, Aloisi AL, Bonasera V, Lattanzio R, de LR, Weidle UH, Piantelli M, Alberti S. Upregulation of Trop-2 quantitatively stimulates human cancer growth. Oncogene. 2013;32:222–233.
    1. Guerra E, Trerotola M, Dell AR, Bonasera V, Palombo B, El-Sewedy T, Ciccimarra T, Crescenzi C, Lorenzini F, Rossi C, Vacca G, Lattanzio R, Piantelli M, Alberti S. A bicistronic CYCLIN D1-TROP2 mRNA chimera demonstrates a novel oncogenic mechanism in human cancer. Cancer Res. 2008;68:8113–8121.
    1. Bignotti E, Todeschini P, Calza S, Falchetti M, Ravanini M, Tassi RA, Ravaggi A, Bandiera E, Romani C, Zanotti L, Tognon G, Odicino FE, Facchetti F, Pecorelli S, Santin AD. Trop-2 overexpression as an independent marker for poor overall survival in ovarian carcinoma patients. Eur J Cancer. 2010;46:944–953.
    1. Fang YJ, Lu ZH, Wang GQ, Pan ZZ, Zhou ZW, Yun JP, Zhang MF, Wan DS. Elevated expressions of MMP7, TROP2, and survivin are associated with survival, disease recurrence, and liver metastasis of colon cancer. Int J Colorectal Dis. 2009;24:875–884.
    1. Fong D, Spizzo G, Gostner JM, Gastl G, Moser P, Krammel C, Gerhard S, Rasse M, Laimer K. TROP2: a novel prognostic marker in squamous cell carcinoma of the oral cavity. Mod Pathol. 2008;21:186–191.
    1. Muhlmann G, Spizzo G, Gostner J, Zitt M, Maier H, Moser P, Gastl G, Zitt M, Muller HM, Margreiter R, Ofner D, Fong D. TROP2 expression as prognostic marker for gastric carcinoma. J Clin Pathol. 2009;62:152–158.
    1. Nakashima K, Shimada H, Ochiai T, Kuboshima M, Kuroiwa N, Okazumi S, Matsubara H, Nomura F, Takiguchi M, Hiwasa T. Serological identification of TROP2 by recombinant cDNA expression cloning using sera of patients with esophageal squamous cell carcinoma. Int J Cancer. 2004;112:1029–1035.
    1. Ambrogi F, Fornili M, Boracchi P, Trerotola M, Relli V, Simeone P, La SR, Lattanzio R, Querzoli P, Pedriali M, Piantelli M, Biganzoli E, Alberti S. Trop-2 is a determinant of breast cancer survival. PLoS One. 2014;9:e96993.
    1. Lin H, Huang JF, Qiu JR, Zhang HL, Tang XJ, Li H, Wang CJ, Wang ZC, Feng ZQ, Zhu J. Significantly upregulated TACSTD2 and Cyclin D1 correlate with poor prognosis of invasive ductal breast cancer. Exp Mol Pathol. 2013;94:73–78.
    1. Wu M, Liu L, Chan C. Identification of novel targets for breast cancer by exploring gene switches on a genome scale. BMC Genomics. 2011;12:547.
    1. Govindan SV, Stein R, Qu Z, Chen S, Andrews P, Ma H, Hansen HJ, Griffiths GL, Horak ID, Goldenberg DM. Preclinical therapy of breast cancer with a radioiodinated humanized anti-EGP-1 monoclonal antibody: advantage of a residualizing iodine radiolabel. Breast Cancer Res Treat. 2004;84:173–182.
    1. Starodub AN, Ocean AO, Shah MA, Guarino MJ, Picozzi VJ, Vahdat LT, Thomas SS, Govindan SV, Maliakla PP, Wegener WA, Hamburger SA, Sharkey RM, Goldenberg DM. First-in-human trial of a novel anti-Trop-2 antibody-SN-38 conjugate, sacituzumab govitecan, for the treatment of diverse metastatic solid tumors. Clin Cancer Res. 2015 May;:Pii. clincancre.3321.2014 (in press)
    1. Bardia A, Starodub A, Moroose RL, Mayer IA, Diamond JR, Chuang E, Govindan SV, Sharkey RM, Maliakal PP, Wegener WA, Hamburger SA, Ocean AO, Goldenberg DM, Vahdat LT. IMMU-132, a new antibody-drug conjugate (ADC) against Trop-2, as a novel therapeutic for patients with relapsed/refractory, metastatic, triple-negative breast cancer (TNBC): Results from Phase I/II clinical trial (NCT01631552) San Antonio Breast Cancer Symposium. 2014;p5:19–27.
    1. Garcia-Carbonero R, Supko JG. Current perspectives on the clinical experience, pharmacology, and continued development of the camptothecins. Clin Cancer Res. 2002;8:641–661.
    1. Kawato Y, Aonuma M, Hirota Y, Kuga H, Sato K. Intracellular roles of SN-38, a metabolite of the camptothecin derivative CPT-11, in the antitumor effect of CPT-11. Cancer Res. 1991;51:4187–4191.
    1. Burke TG, Mi Z. The structural basis of camptothecin interactions with human serum albumin: impact on drug stability. J Med Chem. 1994;37:40–46.
    1. Giovanella BC, Harris N, Mendoza J, Cao Z, Liehr J, Stehlin JS. Dependence of anticancer activity of camptothecins on maintaining their lactone function. Ann NY Acad Sci. 2000;922:27–35.
    1. Moon SJ, Govindan SV, Cardillo TM, D'Souza CA, Hansen HJ, Goldenberg DM. Antibody conjugates of 7-ethyl-10-hydroxycamptothecin (SN-38) for targeted cancer chemotherapy. J Med Chem. 2008;51:6916–6926.
    1. Govindan SV, Cardillo TM, Moon SJ, Hansen HJ, Goldenberg DM. CEACAM5-targeted therapy of human colonic and pancreatic cancer xenografts with potent labetuzumab-SN-38 immunoconjugates. Clin Cancer Res. 2009;15:6052–6061.
    1. Zhao H, Lee C, Sai P, Choe YH, Boro M, Pendri A, Guan S, Greenwald RB. 20-O-acylcamptothecin derivatives: evidence for lactone stabilization 1. J Org Chem. 2000;65:4601–4606.
    1. Govindan SV, Goldenberg DM. Designing immunoconjugates for cancer therapy. Expert Opin Biol Ther. 2012;12:873–890.
    1. Lambert JM. Drug-conjugated antibodies for the treatment of cancer. Br J Clin Pharmacol. 2013;76:248–262.
    1. Govindan SV, Cardillo TM, Sharkey RM, Tat F, Gold DV, Goldenberg DM. Milatuzumab-SN-38 conjugates for the treatment of CD74+ cancers. Mol Cancer Ther. 2013;12:968–978.
    1. Sharkey RM, Juweid M, Shevitz J, Behr T, Dunn R, Swayne LC, Wong GY, Blumenthal RD, Griffiths GL, Siegel JA. Evaluation of a complementarity-determining region-grafted (humanized) anti-carcinoembryonic antigen monoclonal antibody in preclinical and clinical studies. Cancer Res. 1995;55:5935s–5945s.
    1. Cardillo TM, Govindan SV, Sharkey RM, Trisal P, Arrojo R, Liu D, Rossi EA, Chang CH, Goldenberg DM. Sacituzumab govitecan (IMMU-132), an anti-Trop-2/SN-38 antibody-drug conjugate: Characterization and efficacy in pancreatic, gastric, and other cancers. Bioconjug Chem. 2015;26:919–931.
    1. Bonner WM, Redon CE, Dickey JS, Nakamura AJ, Sedelnikova OA, Solier S, Pommier Y. GammaH2AX and cancer. Nat Rev Cancer. 2008;8:957–967.
    1. Zamboni WC, Stewart CF, Cheshire PJ, Richmond LB, Hanna SK, Luo X, Poquette C, McGovren JP, Houghton JA, Houghton PJ. Studies of the efficacy and pharmacology of irinotecan against human colon tumor xenograft models. Clin Cancer Res. 1998;4:743–753.
    1. Raji R, Guzzo F, Carrara L, Varughese J, Cocco E, Bellone S, Betti M, Todeschini P, Gasparrini S, Ratner E, Silasi DA, Azodi M, Schwartz P, Rutherford TJ, Buza N, Pecorelli S, et al. Uterine and ovarian carcinosarcomas overexpressing Trop-2 are sensitive to hRS7, a humanized anti-Trop-2 antibody. J Exp Clin Cancer Res. 2011;30:106.
    1. Varughese J, Cocco E, Bellone S, Bellone M, Todeschini P, Carrara L, Schwartz PE, Rutherford TJ, Pecorelli S, Santin AD. High-grade, chemotherapy-resistant primary ovarian carcinoma cell lines overexpress human trophoblast cell-surface marker (Trop-2) and are highly sensitive to immunotherapy with hRS7, a humanized monoclonal anti-Trop-2 antibody. Gynecol Oncol. 2011;122:171–177.
    1. Varughese J, Cocco E, Bellone S, Ratner E, Silasi DA, Azodi M, Schwartz PE, Rutherford TJ, Buza N, Pecorelli S, Santin AD. Cervical carcinomas overexpress human trophoblast cell-surface marker (Trop-2) and are highly sensitive to immunotherapy with hRS7, a humanized monoclonal anti-Trop-2 antibody. Am J Obstet Gynecol. 2011;205:567–567.
    1. Truong A, Feng N, Sayegh D, Mak B, Hahn S, Pereira D, Young D. AR47A6.4.2, a naked monoclonal antibody targeting Trop-2, exhibits anti-tumor efficacy in multiple human cancer models as a monotherapeutic agent and demonstrates efficacy in combination therapy. Cancer Res. 2008;68 abst 3990.
    1. Lin H, Zhang H, Wang J, Lu M, Zheng F, Wang C, Tang X, Xu N, Chen R, Zhang D, Zhao P, Zhu J, Mao Y, Feng Z. A novel human Fab antibody for Trop2 inhibits breast cancer growth in vitro and in vivo. Int J Cancer. 2014;134:1239–1249.
    1. Firer MA, Gellerman G. Targeted drug delivery for cancer therapy: the other side of antibodies. J Hematol Oncol. 2012;5:70.
    1. Sievers EL, Senter PD. Antibody-drug conjugates in cancer therapy. Annu Rev Med. 2013;64:15–29.
    1. Stepan LP, Trueblood ES, Hale K, Babcook J, Borges L, Sutherland CL. Expression of Trop2 cell surface glycoprotein in normal and tumor tissues: potential implications as a cancer therapeutic target. J Histochem Cytochem. 2011;59:701–710.
    1. Hamblett KJ, Senter PD, Chace DF, Sun MM, Lenox J, Cerveny CG, Kissler KM, Bernhardt SX, Kopcha AK, Zabinski RF, Meyer DL, Francisco JA. Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate. Clin Cancer Res. 2004;10:7063–7070.
    1. Rispens T, Davies AM, Ooijevaar-de Heer P, Absalah S, Bende O, Sutton BJ, Vidarsson G, Aalberse RC. Dynamics of inter-heavy chain interactions in human immunoglobulin G (IgG) subclasses studied by kinetic Fab arm exchange. J Biol Chem. 2014;289:6098–6109.
    1. Gupta E, Lestingi TM, Mick R, Ramirez J, Vokes EE, Ratain MJ. Metabolic fate of irinotecan in humans: correlation of glucuronidation with diarrhea 1. Cancer Res. 1994;54:3723–3725.
    1. Xie R, Mathijssen RH, Sparreboom A, Verweij J, Karlsson MO. Clinical pharmacokinetics of irinotecan and its metabolites: a population analysis. J Clin Oncol. 2002;20:3293–3301.
    1. Stein A, Voigt W, Jordan K. Chemotherapy-induced diarrhea: pathophysiology, frequency and guideline-based management. Ther Adv Med Oncol. 2010;2:51–63.
    1. Hirose K, Kozu C, Yamashita K, Maruo E, Kitamura M, Hasegawa J, Omoda K, Murakami T, Maeda Y. Correlation between plasma concentration ratios of SN-8 glucuronide and SN-8 and neutropenia induction in patients with colorectal cancer and wild-type UGT1A1 gene. Oncol Lett. 2012;3:694–698.
    1. Sharkey RM, McBride WJ, Cardillo TM, Govindan SV, Wang Y, Rossi EA, Chang CH, Goldenberg DM. Enhanced delivery of SN-38 to human tumor xenografts with an anti-Trop-2 SN-38 antibody conjugate (sacituzumab govitecan) Clin Cancer Res. 2015 (in press)

Source: PubMed

3
订阅