Cancer Stem Cell Assay for the Treatment of Platinum-Resistant Recurrent Ovarian Cancer

Candace M Howard, Stephen Bush 2nd, Nadim Bou Zgheib, Seth T Lirette, Antonio Cortese, Antonio Mollo, Jagan Valluri, Pier Paolo Claudio, Candace M Howard, Stephen Bush 2nd, Nadim Bou Zgheib, Seth T Lirette, Antonio Cortese, Antonio Mollo, Jagan Valluri, Pier Paolo Claudio

Abstract

Background: Disease recurrence and progression of ovarian cancer is a common event, which is accompanied by the development of platinum-resistant or refractory disease. The presence of chemo-resistant Cancer Stem Cells (CSCs) contribute to tumor propagation, maintenance, and treatment resistance of this difficult to treat disease. We have developed ChemoID, a cytotoxic synergy assay against CSCs that identifies the most effective chemotherapy treatment from a panel of FDA-approved chemotherapies using fresh cancer biopsies.

Patients and methods: Ascites or interventional radiology biopsies were collected under physician order from 78 consecutive patients affected by 3rd relapsed ovarian cancer. Test results from the assay were used when possible to treat patients with the highest cell kill drugs, taking into consideration their health status and using dose reductions, if needed. A chart analysis and review of CT and PET scans were performed to determine patients' outcomes for tumor response, Progression-Free Survival (PFS), and Overall Survival (OS).

Results: We observed that recurrent ovarian cancer patients treated with high-cell kill chemotherapy agents guided by the CSCs drug response assay had an improvement in their median PFS and OS when compared to historical median PFS and OS and/or when compared to patients who could not receive high cell kill chemotherapies (PFS low cell kill 3.5 months vs. high cell kill 12.0 months; OS low cell kill 6.0 months vs. high cell kill 15.0 months).

Conclusion: This data indicates that the drug cytotoxicity assay aimed at targeting CSCs may be a useful tool for optimizing treatment selection when first-line therapy fails, and when there are multiple clinically-acceptable and -equivalent treatments available.

Keywords: Cancer stem cells; ChemoID; Chemotherapy; Ovarian cancer; Personalized medicine; Platinum resistant ovarian cancer.

Conflict of interest statement

9. Conflict of Interest Authors P.P.C. and J.V. hold intellectual property rights on the use of the ChemoID cancer stem cell assay. All other authors have no relevant disclosures to declare.

Figures

Figure 1:
Figure 1:
Limiting dilution assay of patient-derived CSC and non-CSC injected intraperitoneally in nude mice. Arrows point tumors formed following injections of CSC sorted from biopsies of ovarian cancer. A) Intraperitoneal tumors formed following injection of 1×10^2 CD44(+) CSCs. B) Intraperitoneal tumors formed following injection of 1×10^2 CD133(+) CSCs. C) Intraperitoneal tumors formed following injection of 1×10^2 CD117(+) CSCs. D) Intraperitoneal tumors formed following an injection of 1×10^2 ChemoID enriched CSCs. E) Single intraperitoneal tumor formed, as evidenced by the arrow, following injection of 1×10^6 non-CSCs (negative for CD44, CD133, or CD117) sorted from a biopsy obtained from a patient affected by ovarian cancer.
Figure 2:
Figure 2:
Kaplan Meier analysis of progression-free survival (PFS) of 78 real-world recurrent ovarian cancer patients treated with ChemoID-guided therapy.
Figure 3:
Figure 3:
Kaplan Meier analysis of Progression-Free Survival (PFS) of 78 real-world recurrent ovarian cancer patients stratified by responding drugs vs. non responding drugs according to the percentage of cell kill on CSC and Bulk of Tumor found by the ChemoID assay.
Figure 4:
Figure 4:
Patients are represented as red empty circles who manifested a recurrence of their ovarian cancer within 6-months from therapy start or as blue circles who didn’t manifest recurrence in the same period of time. % cell kill cut-offs were identified for CSCs (40%) and bulk of tumor (55%).
Figure 5:
Figure 5:
Patients are represented as red empty circles who manifested a recurrence of their ovarian cancer within 9-months from therapy start or as blue circles who didn’t manifest recurrence in the same period of time. % cell kill cut-offs were identified for CSCs (40%) and bulk of tumor (55%).
Figure 6:
Figure 6:
Patients are represented as red empty circles who manifested a recurrence of their ovarian cancer within 12-months from therapy start or as blue circles who didn’t manifest recurrence in the same period of time. % cell kill cut-offs were identified for CSCs (40%) and bulk of tumor (55%).
Figure 7:
Figure 7:
Patients represented as red empty circles died for recurence of their ovarian cancer within 6-months from therapy start. Patients represented as blue circles were alive at 6-months from therapy start. % cell kill cut-offs were identified for CSCs (40%) and bulk of tumor (55%).
Figure 8:
Figure 8:
Patients represented as red empty circles died for recurrence of their ovarian cancer within 9-months from therapy start. Patients represented as blue circles were alive at 9-months from therapy start. % cell kill cut-offs were identified for CSCs (40%) and bulk of tumor (55%).
Figure 9:
Figure 9:
Patients represented as red empty circles died for recurrence of their ovarian cancer within 12-months from therapy start. Patients represented as blue circles were alive at 12-months from therapy start. % cell kill cut-offs were identified for CSCs (40%) and bulk of tumor (55%).
Figure 10:
Figure 10:
Kaplan Meier analysis of Overall Survival (OS) of 78 real-world recurrent ovarian cancer patients treated with ChemoID–guided therapy.
Figure 11:
Figure 11:
Kaplan Meier analysis of Overall Survival (OS) of 78 real-world recurrent ovarian cancer patients stratified by responding drugs vs. non responding drugs according to the percentage of cell kill on CSC and Bulk of Tumor found by the ChemoID assay.

References

    1. Hutchinson L, Romero D (2016) Precision or imprecision medicine? Nat Rev Clin Oncol 13: 713.
    1. Deng X, Nakamura Y (2017) Cancer Precision Medicine: From Cancer Screening to Drug Selection and Personalized Immunotherapy. Trends Pharmacol Sci 38: 15–24.
    1. Millner LM, Strotman LN (2016) The Future of Precision Medicine in Oncology. Clin Lab Med 36: 557–573.
    1. Prasad V (2016) Perspective: The precision-oncology illusion. Nature 537: S63.
    1. Maenpaa JU, Heinonen E, Hinkka SM, Karnani P, Klemi PJ, et al. (1995) The subrenal capsule assay in selecting chemotherapy for ovarian cancer: a prospective randomized trial. Gynecol Oncol 57: 294–298.
    1. Suonio E, Lipponen P, Maenpaa J, Syrjanen K, Kangas L, et al. (1997) Mitotic index in the subrenal capsule assay as an indicator of the chemosensitivity of ovarian cancer. Cancer Chemother Pharmacol 41: 15–21.
    1. Von Hoff DD, Kronmal R, Salmon SE, Turner J, Green JB, et al. (1991) A Southwest Oncology Group study on the use of a human tumor cloning assay for predicting response in patients with ovarian cancer. Cancer 67: 20–27.
    1. Wu B, Zhu JS, Zhang Y, Shen WM, Zhang Q (2008) Predictive value of MTT assay as an in vitro chemosensitivity testing for gastric cancer: one institution’s experience. World J Gastroenterol 14: 3064–3068.
    1. Whitehouse PA, Knight LA, Di Nicolantonio F, Mercer SJ, Sharma S, et al. (2003) Heterogeneity of chemosensitivity of colorectal adenocarcinoma determined by a modified ex vivo ATP-tumor chemosensitivity assay (ATP-TCA). Anticancer Drugs 14: 369–375.
    1. Cree IA, Pazzagli M, Mini E, Mazzei T, Elizabeth MMH, et al. (1995) Methotrexate chemosensitivity by ATP luminescence in human leukemia cell lines and in breast cancer primary cultures: comparison of the TCA-100 assay with a clonogenic assay. Anticancer Drugs 6: 398–404.
    1. Mehta RS, Bornstein R, Yu IR, Parker RJ, McLaren CE, et al. (2001) Breast cancer survival and in vitro tumor response in the extreme drug resistance assay. Breast Cancer Res Treat 66: 225–237.
    1. Shaw GL, Gazdar AF, Phelps R, Steinberg SM, Linnoila RI, et al. (1996) Correlation of in vitro drug sensitivity testing results with response to chemotherapy and survival: comparison of non-small cell lung cancer and small cell lung cancer. J Cell Biochem Suppl 24: 173–185.
    1. Cortazar P, Gazdar AF, Woods E, Russell E, Steinberg SM, et al. (1997) Survival of patients with limited-stage small cell lung cancer treated with individualized chemotherapy selected by in vitro drug sensitivity testing. Clin Cancer Res 3: 741–747.
    1. Dollner R, Granzow C, Helmke BM, Ruess A, Schad A, et al. (2004) The impact of stromal cell contamination on chemosensitivity testing of head and neck carcinoma. Anticancer Res 24: 325–331.
    1. Richard S, Wells A, Connor J, Price F (2005) Use of ChemoFx(R) for Identification of Effective Treatments in Epithelial Ovarian Cancer. PLoS Curr 2015: 7
    1. Grendys EC Jr, Fiorica JV, Orr JW Jr, Holloway R, Wang D, et al. (2014) Overview of a chemoresponse assay in ovarian cancer. Clin Transl Oncol 16: 761–769.
    1. Hoffman RM (1991) In vitro sensitivity assays in cancer: a review, analysis, and prognosis. J Clin Lab Anal 5: 133–143.
    1. Sugihara E, Saya H (2013) Complexity of cancer stem cells. Int J Cancer 132: 1249–1259.
    1. Colak S, Medema JP (2014) Cancer stem cells--important players in tumor therapy resistance. FEBS J 281: 4779–4791.
    1. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, et al. (1994) A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367: 645–648.
    1. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100: 3983–3988.
    1. Islam F, Qiao B, Smith RA, Gopalan V, Lam AK (2005) Cancer stem cell: fundamental experimental pathological concepts and updates. Exp Mol Pathol 98: 184–191.
    1. Landen CN Jr, Goodman B, Katre AA, Steg AD, Nick AM, et al. (2010) Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer. Mol Cancer Ther 9: 3186–3199.
    1. Kleffel S, Schatton T (2013) Tumor dormancy and cancer stem cells: two sides of the same coin? Adv Exp Med Biol 734: 145–179.
    1. Turdo A, Veschi V, Gaggianesi M, Chinnici A, Bianca P, et al. (2019) Meeting the Challenge of Targeting Cancer Stem Cells. Front Cell Dev Biol 7: 16.
    1. Maugeri-Sacca M, Vigneri P, De Maria R (2011) Cancer stem cells and chemosensitivity. Clin Cancer Res 17: 4942–4947.
    1. van Niekerk G, Davids LM, Hattingh SM, Engelbrecht AM (2017) Cancer stem cells: A product of clonal evolution? Int J Cancer 140: 993–999.
    1. Mathis SE, Alberico A, Nande R, Neto R, Neto W, et al. (2014) Chemo-predictive assay for targeting cancer stem-like cells in patients affected by brain tumors. PLoS One 9: e105710.
    1. Pan ST, Li ZL, He ZX, Qiu JX, Zhou SF (2016) Molecular mechanisms for tumor resistance to chemotherapy. Clin Exp Pharmacol Physiol 48: 723–737.
    1. Herzog TJ, Krivak TC, Fader AN, Coleman RL (2010) Chemosensitivity testing with ChemoFx and overall survival in primary ovarian cancer. Am J Obstet Gynecol 203: 68 e61–66.
    1. Ness RB, Wisniewski SR, Eng H, Christopherson W (2002) Cell viability assay for drug testing in ovarian cancer: in vitro kill versus clinical response. Anticancer Res 22: 1145–1149.
    1. Cortese A, Pantaleo G, Amato M, Lawrence L, Mayes V, et al. (2016) A new complementary procedure for patients affected by head and neck cancer: Chemo-predictive assay. Int J Surg Case Rep 26: 42–46.
    1. Kelly SE, Di Benedetto A, Greco A, Howard CM, Sollars VE, et al. (2010) Rapid selection and proliferation of CD133+ cells from cancer cell lines: chemotherapeutic implications. PLoS One 5: e10035.
    1. Ranjan T, Howard CM, Yu A, Xu L, Aziz K, et al. (2020) Cancer Stem Cell Chemotherapeutics Assay for Prospective Treatment of Recurrent Glioblastoma and Progressive Anaplastic Glioma: A Single-Institution Case Series. Transl Oncol 13: 100755.
    1. Howard CM, Valluri J, Alberico A, Julien T, Mazagri R, et al. (2017) Analysis of Chemopredictive Assay for Targeting Cancer Stem Cells in Glioblastoma Patients. Transl Oncol 10: 241–254.
    1. DeEulis T, Howard CM, Valluri J, Claudio PP (2017) Efficacy of ChemoID guided drug selection for palliative chemotherapy in advanced recurrent high-grade ovarian adenocarcinoma: Case Study. Transl Med Rep 1: 70–75.
    1. Howard CM, Zgheib NB, Bush S 2nd, DeEulis T, Cortese A, et al. (2020) Clinical relevance of cancer stem cell chemotherapeutic assay for recurrent ovarian cancer. Transl Oncol 13: 100860.
    1. Howard CM, Valluri J, Claudio PP (2017) Functional drug response assay for cancer stem cells in the era of precision medicine. Translational Medicine Report. 2017: 1.
    1. Hanker LC, Loibl S, Burchardi N, Pfisterer J, Meier W, et al. (2012) The impact of second to sixth line therapy on survival of relapsed ovarian cancer after primary taxane/platinum-based therapy. Ann Oncol 23: 2605–2612.
    1. Ahmed N, Abubaker K, Findlay J, Quinn M (2013) Cancerous ovarian stem cells: obscure targets for therapy but relevant to chemoresistance. J Cell Biochem 114: 21–34.
    1. Ahmed N, Abubaker K, Findlay JK (2014) Ovarian cancer stem cells: Molecular concepts and relevance as therapeutic targets. Mol Aspects Med 39: 110–125.
    1. Bapat SA, Mali AM, Koppikar CB, Kurrey NK (2005) Stem and progenitor-like cells contribute to the aggressive behavior of human epithelial ovarian cancer. Cancer Res 65: 3025–3029.
    1. Burgos-Ojeda D, Rueda BR, Buckanovich RJ (2012) Ovarian cancer stem cell markers: prognostic and therapeutic implications. Cancer Lett 322: 1–7.
    1. Metastatic Renal Cell Cancer: Summary from ASCO 2015. Can Urol Assoc J 9: S158–S161.
    1. Fotopoulou C, Zang R, Gultekin M, Cibula D, Ayhan A, et al. (2013) Value of tertiary cytoreductive surgery in epithelial ovarian cancer: an international multicenter evaluation. Ann Surg Oncol 20: 1348–1354.
    1. Stockler MR, Hilpert F, Friedlander M, King MT, Wenzel L, et al. (2014) Patient-reported outcome results from the open-label phase III AURELIA trial evaluating bevacizumab-containing therapy for platinum-resistant ovarian cancer. J Clin Oncol 32: 1309–1316.
    1. Pujade-Lauraine E, Hilpert F, Weber B, Reuss A, Poveda A, et al. (2014) Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: The AURELIA open-label randomized phase III trial. J Clin Oncol 32: 1302–1308.
    1. Aghajanian C, Blank SV, Goff BA, Judson PL, Teneriello MG, et al. (2012) OCEANS: a randomized, double-blind, placebo-controlled phase III trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent epithelial ovarian, primary peritoneal, or fallopian tube cancer. J Clin Oncol 30: 2039–2045.
    1. Aghajanian C, Goff B, Nycum LR, Wang YV, Husain A, et al. (2015) Final overall survival and safety analysis of OCEANS, a phase 3 trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent ovarian cancer. Gynecol Oncol 139: 10–16.

Source: PubMed

3
Abonner