Safety and efficacy of MIKE-1 in patients with advanced pancreatic cancer: a study protocol for an open-label phase I/II investigator-initiated clinical trial based on a drug repositioning approach that reprograms the tumour stroma

Yasuyuki Mizutani, Tadashi Iida, Eizaburo Ohno, Takuya Ishikawa, Fumie Kinoshita, Yachiyo Kuwatsuka, Miwa Imai, Shinobu Shimizu, Toshihisa Tsuruta, Atsushi Enomoto, Hiroki Kawashima, Mitsuhiro Fujishiro, Yasuyuki Mizutani, Tadashi Iida, Eizaburo Ohno, Takuya Ishikawa, Fumie Kinoshita, Yachiyo Kuwatsuka, Miwa Imai, Shinobu Shimizu, Toshihisa Tsuruta, Atsushi Enomoto, Hiroki Kawashima, Mitsuhiro Fujishiro

Abstract

Background: Cancer-associated fibroblasts (CAFs) are an important component of the tumour microenvironment. Recent studies revealed CAFs are heterogeneous and CAF subset(s) that suppress cancer progression (cancer-restraining CAFs [rCAFs]) must exist in addition to well-characterised cancer-promoting CAFs (pCAFs). However, the identity and specific markers of rCAFs are not yet reported. We recently identified Meflin as a specific marker of rCAFs in pancreatic and colon cancers. Our studies revealed that rCAFs may represent proliferating resident fibroblasts. Interestingly, a lineage tracing experiment showed Meflin-positive rCAFs differentiate into α-smooth muscle actin-positive and Meflin-negative CAFs, which are generally hypothesised as pCAFs, during cancer progression. Using a pharmacological approach, we identified AM80, a synthetic unnatural retinoid, as a reagent that effectively converts Meflin-negative pCAFs to Meflin-positive rCAFs. We aimed to investigate the efficacy of a combination of AM80 and gemcitabine (GEM) and nab-paclitaxel (nab-PTX) in patients with advanced pancreatic cancer.

Methods: The phase I part is a 3 + 3 design, open-label, and dose-finding study. The dose-limiting toxicity (DLT) of these combination therapies would be evaluated for 4 weeks. After the DLT evaluation period, if no disease progression is noted based on the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 or if the patient has no intolerable toxicity, administration of AM80 with GEM and nab-PTX would be continued for up to 24 weeks. The phase II part is an open-label, single-arm study. The maximum tolerated dose (MTD) of AM80 with GEM and nab-PTX, determined in phase I, would be administered until intolerable toxicity or disease progression occurs, up to a maximum of 24 weeks, to confirm efficacy and safety. The primary endpoints are frequency of DLT and MTD of AM80 with GEM and nab-PTX in the phase I part and response rate based on the RECIST in the phase II part. Given the historical control data, we hope that the response rate will be over 23% in phase II.

Discussion: Strategies to convert pCAFs into rCAFs have been developed in recent years. We hypothesised that AM80 would be a promising enhancer of chemosensitivity and drug distribution through CAF conversion in the stroma.

Trial registration: Clinicaltrial.gov: NCT05064618 , registered on 1 October 2021. jRCT: jRCT2041210056 , registered on 27 August 2021.

Keywords: AM80; Cancer stroma; Cancer-associated fibroblasts; Cancer-restraining CAFs; ISLR; MIKE-1; Meflin; Tamibarotene; Tumour microenvironment.

Conflict of interest statement

In implementing this study, the investigators and sub-investigators are provided with no funds or services by the manufacturer of the investigational drug or the organisation that tests drug levels. This study will be implemented after being reviewed for conflicts of interest by the institutional review board of our hospital under appropriate management. As for a conflict of interest that may occur in this study, any related persons including the investigators may obtain a reward in the future as inventors of a patent to be filed that is related to this therapy, which will be handled according to the rules of Nagoya University Hospital.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Representative histology of human pancreatic ductal adenocarcinoma (PDAC) and diversity of cancer-associated fibroblasts (CAFs). The representative histology of human PDAC is shown in the left panel. Note that the volume of the tumour stroma is predominant in cancer cells. In tumour stroma, there are many proliferating CAFs that deposit a large amount of the extracellular matrix. CAFs can be classified based on their functions into cancer-promoting CAFs and cancer-restraining (rCAFs). Meflin is a marker of rCAF and inhibits tissue fibrosis by augmenting bone morphogenetic protein 7 signalling and inhibiting lysyl oxidase activity. AM80 may induce the reprogramming of tumour stroma by upregulating the expression of Meflin in CAFs
Fig. 2
Fig. 2
Protocol of the present study. In the present study, AM80 will be administered daily for 4 weeks and repeated for up to six courses. Each course consists of gemcitabine (GEM) (1,000 mg/m2) and nab-paclitaxel (nab-PTX) (125 mg/m2) administered intravenously over 30 min on Days 1, 8, and 15 without administration at Week 4. After completing Course 6, if no disease progression is noted based on the Response Evaluation Criteria in Solid Tumors version 1.1 or if the patient has no intolerable toxicity, GEM + nab-PTX will be continued as a usual treatment. In such a case, continuous GEM/nab-PTX will be considered as post-treatment. *Dose-limiting toxicity will be evaluated in a 4-week period only in the phase I trial. **The follow-up period will be set in all cases until the date of completion of the post-observation period (cut-off) in all cases
Fig. 3
Fig. 3
Dose escalation in the phase I part. The AM80 dose will not be reduced or escalated in the same subject. Each course consists of gemcitabine (1,000 mg/m2) and nab-paclitaxel (125 mg/m2) administered intravenously over 30 min on Days 1, 8, and 15, without administration at Week 4. The course will be repeated, and the dose will be reduced ad libitum based on the subject’s condition in compliance with the criteria of this protocol
Fig. 4
Fig. 4
Flowcharts of steps to determine dose-limiting toxicities in the phase I study. In the phase 1 part study, this is a dose-escalation study using a standard 3 + 3 design. See text for details

References

    1. Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21:418–429. doi: 10.1016/j.ccr.2012.01.007.
    1. Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013;62:112–120. doi: 10.1136/gutjnl-2012-302529.
    1. Sherman MH, Yu RT, Engle DD, Ding N, Atkins AR, Tiriac H, et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell. 2014;159:80–93. doi: 10.1016/j.cell.2014.08.007.
    1. Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–747. doi: 10.1016/j.ccr.2014.04.021.
    1. Shin K, Lim A, Zhao C, Sahoo D, Pan Y, Spiekerkoetter E, et al. Hedgehog signaling restrains bladder cancer progression by eliciting stromal production of urothelial differentiation factors. Cancer Cell. 2014;26:521–533. doi: 10.1016/j.ccell.2014.09.001.
    1. Özdemir BC, Pentcheva-Hoang T, Carstens J, Zheng X, Wu C, Simpson T, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25:719–734. doi: 10.1016/j.ccr.2014.04.005.
    1. Neesse A, Bauer CA, Öhlund D, Lauth M, Buchholz M, Michl P, et al. Stromal biology and therapy in pancreatic cancer: ready for clinical translation? Gut. 2019;68:159–171. doi: 10.1136/gutjnl-2018-316451.
    1. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16:582–598. doi: 10.1038/nrc.2016.73.
    1. Mizutani Y, Kobayashi H, Iida T, Asai N, Masamune A, Hara A, et al. Meflin-positive cancer-associated fibroblasts inhibit pancreatic carcinogenesis. Cancer Res. 2019;79:5367–5381. doi: 10.1158/0008-5472.CAN-19-0454.
    1. Kobayashi H, Enomoto A, Woods SL, Burt AD, Takahashi M, Worthley DL. Cancer-associated fibroblasts in gastrointestinal cancer. Nat Rev Gastroenterol Hepatol. 2019;16:282–295. doi: 10.1038/s41575-019-0115-0.
    1. Takahashi M, Kobayashi H, Mizutani Y, Hara A, Iida T, Miyai Y, et al. Roles of the mesenchymal Stromal/Stem cell Marker Meflin/Islr in Cancer and Fibrosis. Front Cell Dev Biol. 2021;9:749924. doi: 10.3389/fcell.2021.749924.
    1. Kobayashi H, Gieniec KA, Wright JA, Wang T, Asai N, Mizutani Y, et al. The balance of stromal BMP signaling mediated by GREM1 and ISLR drives colorectal carcinogenesis. Gastroenterology. 2021;160:1224–39.e30. doi: 10.1053/j.gastro.2020.11.011.
    1. Maeda K, Enomoto A, Hara A, Asai N, Kobayashi T, Horinouchi A, et al. Identification of Meflin as a potential marker for mesenchymal stromal cells. Sci Rep. 2016;6:22288. doi: 10.1038/srep22288.
    1. Hara A, Kobayashi H, Asai N, Saito S, Higuchi T, Kato K, et al. Roles of the mesenchymal stromal/stem cell marker Meflin in cardiac tissue repair and the development of diastolic dysfunction. Circ Res. 2019;125:414–430. doi: 10.1161/CIRCRESAHA.119.314806.
    1. Iida T, Mizutani Y, Esaki N, Ponic SM, Burkel BM, Weng R, et al. Conversion of cancer-associated fibroblasts from pro- to antitumor improves the sensitivity of pancreatic cancer to chemotherapeutics. bioRxiv. 2021. Accessed [18 Nov 2021] 10.1101/2021.06.29.450327.
    1. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020;20:174–186. doi: 10.1038/s41568-019-0238-1.
    1. Chen Y, McAndrews KM, Kalluri R, et al. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 2021;18:792–804. doi: 10.1038/s41571-021-00546-5.
    1. Umemiya H, Fukasawa H, Ebisawa M, Eyrolles L, Kawachi E, Eisenmann G, et al. Regulation of retinoidal actions by diazepinylbenzoic acids. Retinoid synergists which activate the RXR-RAR heterodimers. J Med Chem. 1997;40:4222–34. doi: 10.1021/jm9704309.
    1. Kagechika H, Kawachi E, Hashimoto Y, Himi T, Shudo K. Retinobenzoic acids. 1. Structure-activity relationships of aromatic amides with retinoidal activity. J Med Chem. 1988;31:2182–92. doi: 10.1021/jm00119a021.
    1. Tobita T, Takeshita A, Kitamura K, Ohnishi K, Yanagi M, Hiraoka A, et al. Treatment with a new synthetic retinoid, Am 80, of acute promyelocytic leukemia relapsed from complete remission induced by all-trans retinoic acid. Blood. 1997;90:967–973. doi: 10.1182/blood.V90.3.967.
    1. Classification of Pancreatic Carcinoma 4th English Edition. Japan Pancreas Society. Accessed [18 Nov 2021]. .
    1. Arrieta O, González-De la Rosa CH, Aréchaga-Ocampo E, Villanueva-Rodríguez G, Cerón-Lizárraga TL, Martínez-Barrera L, et al. Randomized phase II trial of All-trans-retinoic acid with chemotherapy based on paclitaxel and cisplatin as first-line treatment in patients with advanced non-small-cell lung cancer. J Clin Oncol. 2010;28:3463–71. doi: 10.1200/JCO.2009.26.6452.
    1. Kanai F, Obi S, Fujiyama S, Shiina S, Tamai H, Mochizuki H, et al. An open-label Phase I/II study of tamibarotene in patients With advanced hepatocellular carcinoma. Hepatol Int. 2014;8:94–103. doi: 10.1007/s12072-013-9459-7.
    1. Kocher HM, Basu B, Froeling FEM, Sarker D, Slater S, Carlin D, et al. Phase I clinical trial repurposing all-trans retinoic acid as a stromal targeting agent for pancreatic cancer. Nat Commun. 2020;11:4841. doi: 10.1038/s41467-020-18636-w.

Source: PubMed

3
Sottoscrivi