Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kuan-Chun Huang is active.

Publication


Featured researches published by Kuan-Chun Huang.


Molecular Cancer Therapeutics | 2014

Selective Inhibition of EZH2 by EPZ-6438 Leads to Potent Antitumor Activity in EZH2-Mutant Non-Hodgkin Lymphoma

Sarah K. Knutson; Satoshi Kawano; Yukinori Minoshima; Natalie Warholic; Kuan-Chun Huang; Yonghong Xiao; Tadashi Kadowaki; Mai Uesugi; Galina Kuznetsov; Namita Kumar; Tim J. Wigle; Christine R. Klaus; Christina J. Allain; Alejandra Raimondi; Nigel J. Waters; Jesse J. Smith; Margaret Porter-Scott; Richard Chesworth; Mikel P. Moyer; Robert A. Copeland; Victoria M. Richon; Toshimitsu Uenaka; Roy M. Pollock; Kevin Wayne Kuntz; Akira Yokoi; Heike Keilhack

Mutations within the catalytic domain of the histone methyltransferase EZH2 have been identified in subsets of patients with non-Hodgkin lymphoma (NHL). These genetic alterations are hypothesized to confer an oncogenic dependency on EZH2 enzymatic activity in these cancers. We have previously reported the discovery of EPZ005678 and EPZ-6438, potent and selective S-adenosyl-methionine-competitive small molecule inhibitors of EZH2. Although both compounds are similar with respect to their mechanism of action and selectivity, EPZ-6438 possesses superior potency and drug-like properties, including good oral bioavailability in animals. Here, we characterize the activity of EPZ-6438 in preclinical models of NHL. EPZ-6438 selectively inhibits intracellular lysine 27 of histone H3 (H3K27) methylation in a concentration- and time-dependent manner in both EZH2 wild-type and mutant lymphoma cells. Inhibition of H3K27 trimethylation (H3K27Me3) leads to selective cell killing of human lymphoma cell lines bearing EZH2 catalytic domain point mutations. Treatment of EZH2-mutant NHL xenograft-bearing mice with EPZ-6438 causes dose-dependent tumor growth inhibition, including complete and sustained tumor regressions with correlative diminution of H3K27Me3 levels in tumors and selected normal tissues. Mice dosed orally with EPZ-6438 for 28 days remained tumor free for up to 63 days after stopping compound treatment in two EZH2-mutant xenograft models. These data confirm the dependency of EZH2-mutant NHL on EZH2 activity and portend the utility of EPZ-6438 as a potential treatment for these genetically defined cancers. Mol Cancer Ther; 13(4); 842–54. ©2014 AACR.


Oncotarget | 2015

E7449: A dual inhibitor of PARP1/2 and tankyrase1/2 inhibits growth of DNA repair deficient tumors and antagonizes Wnt signaling.

Sharon McGonigle; Zhihong Chen; Jiayi Wu; Paul Chang; Donna Kolber-Simonds; Karen Ackermann; Natalie C. Twine; Jue-lon Shie; Jingzang Tao Miu; Kuan-Chun Huang; George A. Moniz; Kenichi Nomoto

Inhibition of Poly(ADP-ribose) Polymerase1 (PARP1) impairs DNA damage repair, and early generation PARP1/2 inhibitors (olaparib, niraparib, etc.) have demonstrated clinical proof of concept for cancer treatment. Here, we describe the development of the novel PARP inhibitor E7449, a potent PARP1/2 inhibitor that also inhibits PARP5a/5b, otherwise known as tankyrase1 and 2 (TNKS1 and 2), important regulators of canonical Wnt/β-catenin signaling. E7449 inhibits PARP enzymatic activity and additionally traps PARP1 onto damaged DNA; a mechanism previously shown to augment cytotoxicity. Cells deficient in DNA repair pathways beyond homologous recombination were sensitive to E7449 treatment. Chemotherapy was potentiated by E7449 and single agent had significant antitumor activity in BRCA-deficient xenografts. Additionally, E7449 inhibited Wnt/β-catenin signaling in colon cancer cell lines, likely through TNKS inhibition. Consistent with this possibility, E7449 stabilized axin and TNKS proteins resulting in β-catenin de-stabilization and significantly altered expression of Wnt target genes. Notably, hair growth mediated by Wnt signaling was inhibited by E7449. A pharmacodynamic effect of E7449 on Wnt target genes was observed in tumors, although E7449 lacked single agent antitumor activity in vivo, a finding typical for selective TNKS inhibitors. E7449 antitumor activity was increased through combination with MEK inhibition. Particularly noteworthy was the lack of toxicity, most significantly the lack of intestinal toxicity reported for other TNKS inhibitors. E7449 represents a novel dual PARP1/2 and TNKS1/2 inhibitor which has the advantage of targeting Wnt/β-catenin signaling addicted tumors. E7449 is currently in early clinical development.


Molecular Cancer Therapeutics | 2016

Apratoxin A Shows Novel Pancreas-Targeting Activity through the Binding of Sec 61.

Kuan-Chun Huang; Zhihong Chen; Yimin Jiang; Sandeep Akare; Donna Kolber-Simonds; Krista Condon; Sergei Agoulnik; Karen TenDyke; Yongchun Shen; Kuo-Ming Wu; Steven Mathieu; Hyeong-wook Choi; Xiaojie Zhu; Hajime Shimizu; Yoshihiko Kotake; William H. Gerwick; Toshimitsu Uenaka; Mary Woodall-Jappe; Kenichi Nomoto

Apratoxin A is a natural product with potent antiproliferative activity against many human cancer cell lines. However, we and other investigators observed that it has a narrow therapeutic window in vivo. Previous mechanistic studies have suggested its involvement in the secretory pathway as well as the process of chaperone-mediated autophagy. Still the link between the biologic activities of apratoxin A and its in vivo toxicity has remained largely unknown. A better understanding of this relationship is critically important for any further development of apratoxin A as an anticancer drug. Here, we describe a detailed pathologic analysis that revealed a specific pancreas-targeting activity of apratoxin A, such that severe pancreatic atrophy was observed in apratoxin A–treated animals. Follow-up tissue distribution studies further uncovered a unique drug distribution profile for apratoxin A, showing high drug exposure in pancreas and salivary gland. It has been shown previously that apratoxin A inhibits the protein secretory pathway by preventing cotranslational translocation. However, the molecule targeted by apratoxin A in this pathway has not been well defined. By using a 3H-labeled apratoxin A probe and specific Sec 61α/β antibodies, we identified that the Sec 61 complex is the molecular target of apratoxin A. We conclude that apratoxin A in vivo toxicity is likely caused by pancreas atrophy due to high apratoxin A exposure. Mol Cancer Ther; 15(6); 1208–16. ©2016 AACR.


OncoImmunology | 2017

EP4 Antagonism by E7046 diminishes Myeloid immunosuppression and synergizes with Treg-reducing IL-2-Diphtheria toxin fusion protein in restoring anti-tumor immunity

Diana I. Albu; Zichun Wang; Kuan-Chun Huang; Jiayi Wu; Natalie C. Twine; Sarah Leacu; Christy Ingersoll; Lana Parent; Winnie Lee; Diana Liu; Renee Wright-Michaud; Namita Kumar; Galina Kuznetsov; Qian Chen; Wanjun Zheng; Kenichi Nomoto; Mary Woodall-Jappe; Xingfeng Bao

ABSTRACT Reprogramming of immunosuppressive tumor microenvironment (TME) by targeting alternatively activated tumor associated macrophages (M2TAM), myeloid-derived suppressor cells (MDSC), and regulatory T cells (Tregs), represents a promising strategy for developing novel cancer immunotherapy. Prostaglandin E2 (PGE2), an arachidonic acid pathway metabolite and mediator of chronic inflammation, has emerged as a powerful immunosuppressor in the TME through engagement with one or more of its 4 receptors (EP1-EP4). We have developed E7046, an orally bioavailable EP4-specific antagonist and show here that E7046 has specific and potent inhibitory activity on PGE2-mediated pro-tumor myeloid cell differentiation and activation. E7046 treatment reduced the growth or even rejected established tumors in vivo in a manner dependent on both myeloid and CD8+ T cells. Furthermore, co-administration of E7046 and E7777, an IL-2-diphtheria toxin fusion protein that preferentially kills Tregs, synergistically disrupted the myeloid and Treg immunosuppressive networks, resulting in effective and durable anti-tumor immune responses in mouse tumor models. In the TME, E7046 and E7777 markedly increased ratios of CD8+granzymeB+ cytotoxic T cells (CTLs)/live Tregs and of M1-like/M2TAM, and converted a chronic inflammation phenotype into acute inflammation, shown by substantial induction of STAT1/IRF-1 and IFNγ-controlled genes. Notably, E7046 also showed synergistic anti-tumor activity when combined with anti-CTLA-4 antibodies, which have been reported to diminish intratumoral Tregs. Our studies thus reveal a specific myeloid cell differentiation-modifying activity by EP4 blockade and a novel combination of E7046 and E7777 as a means to synergistically mitigate both myeloid and Treg-derived immunosuppression for cancer treatment in preclinical models.


Cancer Research | 2015

Abstract 275: ER-886046, an antagonist of PGE2 receptor type-4, induces an effective antitumor immune response in mice by attenuating intratumoral MDSCs and TAMs

Diana I. Albu; Zichun Wang; Jiayi Wu; Kuan-Chun Huang; Wei Li; Diana Liu; Galina Kuznetsov; Qian Chen; Xingfeng Bao; Mary Woodall-Jappe

One of the hallmarks of an immunosuppressive tumor microenvironment is the presence of myeloid-derived suppressor cells (MDSCs) and type 2 tumor-associated macrophages (TAMs). These myeloid cells derive from immature monocytes and their differentiation is highly regulated by the engagement of prostaglandin E2 (PGE2) receptor type 4 (EP4) with PGE2 in the tumor microenvironment. To understand the importance of EP4 for tumor support, we implanted mouse tumors in EP4 inducible knockout mice and found that these tumors grew significantly more slowly than in wild type mice, indicating an important role of host cell EP4 signaling for tumor progression. Here we report the development and evaluation of ER-886046, a novel and specific EP4 antagonist, for cancer treatment. Daily oral administration of ER-886046 inhibited the growth of multiple mouse syngeneic tumor models with an inhibitory activity up to 100%. The anti-tumor activity of ER-886046 was T cell-dependent since it was not observed in T cell deficient mice. Furthermore, we found in vitro and in vivo mechanistic evidence that ER-886046 interferes with tumor-induced monocyte differentiation into immunosuppressive type 2 macrophages and MDSCs, instead supporting monocyte differentiation into APCs and facilitating intratumoral T cell accumulation. Importantly, ER-886046 has a desirable pharmacokinetic and metabolism profile in mice, rats and dogs which qualifies it as a good candidate for clinical studies in humans. Thus, the preclinical data support further investigation of targeting EP4 signaling by ER-886046 as a novel immune therapy for cancer treatment in clinical setting. Citation Format: Diana I. Albu, Zichun Wang, Jiayi Wu, Kuan-chun Huang, Wei Li, Diana Liu, Galina Kuznetsov, Qian Chen, Xingfeng Bao, Mary Woodall-Jappe. ER-886046, an antagonist of PGE2 receptor type-4, induces an effective antitumor immune response in mice by attenuating intratumoral MDSCs and TAMs. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 275. doi:10.1158/1538-7445.AM2015-275


Journal for ImmunoTherapy of Cancer | 2015

Combination of EP4 antagonist and checkpoint inhibitors promotes anti-tumor effector T cells in preclinical tumor models

Xingfeng Bao; Diana I. Albu; Kuan-Chun Huang; Jiayi Wu; Natalie C. Twine; Kenichi Nomoto; Mary Woodall-Jappe

Immunotherapies targeting the immune checkpoint receptors have shown great promise for a subset of cancer patients. However, robust and safe combination therapies are still needed to increase the benefit of cancer immunotherapy and bring it to broader patient populations. We have recently shown that E7046, a specific EP4 antagonist, possesses significant anti-tumor growth activity in multiple preclinical tumor models through modulating myeloid cells including tumor associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) (AACR 2015, poster #275). Here we evaluated the anti-tumor activities of E7046 in combination with the checkpoint inhibitors anti-CTLA4 and anti-PD1 antibodies, and also with E7777, a recombinant IL-2/diphtheria toxin fusion protein, in immunogenic CT26 and poorly immunogenic 4T1 tumor models. In the CT26 model, concomitant treatment of E7046 and anti-PD1 led to pronounced tumor growth inhibition, with 40% of the mice rendered stably tumor free, while either single agent produced mostly tumor growth inhibitory activity and only an occasional tumor-free animal. In the same model, markedly improved anti-tumor activity was observed for the combination of E7046 and E7777, with up to 20% of animals rendered stably tumor free, compared with only modest anti-tumor activity of either single agent treatment alone. In the 4T1 model, combining E7046 and anti-CTLA4 resulted in a nearly complete tumor growth inhibition; either agent alone had only modest growth inhibitory activity. In the tumor microenvironment of both models, an effective anti-tumor immune response was induced by the combination treatments including E7046 as indicated by the robust accumulation and activation of CD8 cytotoxic T cells (CTL) and/or significantly improved ratio of activated GZMB+CD8+ CTL vs CD4+CD25+Foxp3+ Treg cells. In addition, combination treatments including E7046 in tumor-bearing mice showed no additional gross toxicity compared with immune checkpoint inhibitors alone or E7777 alone. Taken together, these results demonstrated a superior activity of combinational therapies including E7046 over immune checkpoint inhibitors or E7777 alone with acceptable toxicity in preclinical models, and therefore candidates for combination trials in patients. IND filing number of E7046 is 125272.


Cancer Research | 2017

Abstract 4586: Intratumoral Treg cell depletion by local administration of IL-2-Diphteria toxin fusion protein E7777 induces a therapeutic and memory anti-tumor immune response in preclinical models

Diana I. Albu; Christy Ingersoll; Kuan-Chun Huang; Mary Woodall-Jappe; Xingfeng Bao

T regulatory (Treg) cells play an important role in maintaining immunological tolerance to self-antigens, thus limiting immune responses to tumor antigens. Therefore, depleting or suppressing Tregs is one strategy by which anti-tumor immunity can be restored. The immunotoxin ONTAK® is an IL-2-diphteria toxin fusion protein that has been shown to diminish Tregs in patients and animal models of cancer in peripheral blood using a systemic intravenous (i.v.) administration route. E7777 is a new version of ONTAK®. In this study we tested the hypothesis that locally-diminished Tregs by intratumoral (i.t.) administration of E7777 generate effective anti-tumor immune response at both local and systemic levels. First, we showed superior anti-tumor activity and safety of E7777 i.t. over E7777 i.v., where i.t. administration resulted in complete tumor regressions in both moderately immunogenic CT26 and non-immunogenic B16F10 tumors with minimal animal body weight loss. In contrast, only tumor growth delay was observed for E7777 i.v. with dose-limiting animal body weight reduction in the same models. Immune phenotyping showed a 4 fold reduction of intratumoral Tregs in treated CT-26 tumors without significant change of Tregs in the spleens of treated animals, confirming a local Treg-depleting effect of E7777 i.t. In contrast, intratumoral CD8+ T cells were not reduced. Second, E7777 i.t. enhanced overall anti-tumor immune response, manifested by significantly increased numbers of CD45+ hematopoietic cells, Granzyme B+ CD8+ cytotoxic T cells, and ratios of cytotoxic T cells/Tregs in the treated tumors. Importantly, E7777 i.t. also resulted in distant effects in the spleen characterized by increased ratio of T lymphocytes to myeloid cells and increased frequencies of both effector memory CD8+ T cells (CD8+CD62L-CD44+) and central memory CD8+ T cells (CD8+CD62L+CD44+) indicative of systemic immune activation. Consistent with the generation of immunological memory, 60% of the tumor-free animals treated with E7777 i.t. rejected completely and 40% displayed delayed tumor growth of B16F10 cell challenge while all naive control animals grew tumors. Taken together, our results demonstrate that intratumoral Treg depletion by local administration of E7777 leads to an effective local and memory anti-tumor response in preclinical models and support further evaluation of local E7777 delivery as a cancer immunotherapy. Citation Format: Diana I. Albu, Christy Ingersoll, Kuan-Chun Huang, Mary Woodall-Jappe, Xingfeng Bao. Intratumoral Treg cell depletion by local administration of IL-2-Diphteria toxin fusion protein E7777 induces a therapeutic and memory anti-tumor immune response in preclinical models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4586. doi:10.1158/1538-7445.AM2017-4586


Cancer Research | 2017

Abstract 4607: Specific inhibition of PGE2-EP4 signaling by E7046 promotes anti-tumor activity of checkpoint blockade agents through boosting cytotoxic T cell activity

Diana I. Albu; David Verbel; Yuan Huang; Donna Kolber-Simonds; Zichun Wang; Xulong Wang; Zoltan Dezso; Christy Ingersoll; Kuan-Chun Huang; Janna Hutz; Mary Woodall-Jappe; Xingfeng Bao

Purpose: Immunotherapies targeting immune checkpoint receptors have shown great promise for a subset of cancer patients; however, robust and safe combination therapies are still needed. In the tumor microenvironment, prostaglandin E2 receptor type 4 (EP4) signaling has been implicated in both protumoral myeloid cell differentiation and cytotoxic T cell exhaustion. We evaluated the combination of the EP4 antagonist E7046 (clinical trial NCT02540291) with anti-PD1 or anti-CTLA4 in preclinical tumor models, and also interrogated the relationship between PGE2 pathway activation and cancer patient survival. Materials/Methods: Mouse syngeneic tumor models CT-26 and 4T1 were used for pharmacological investigation. GMP grade E7046 was administered to tumor-bearing animals by oral gavage. Co-culture of EG7-OVA and OT1 cells in an antigen-specific cytotoxic T cell (CTL) activation assay provided mechanistic insights. For translational validation, transcripts of five major genes involved in PGE2 synthesis, transport and degradation were compared between malignant and normal tissues across all TCGA tumor types, and correlation of their expression with overall survival was assessed. Results: In the CT26 tumor model, the combination of E7046 and anti-PD1 resulted in significantly more tumor-free animals compared with either agent alone. In the 4T1 tumor model, the combination of E7046 and anti-CTLA4 was also more effective in suppressing tumor growth and tumor rejection compared with anti-CTLA4 alone, and was accompanied by a markedly increased accumulation of GZMB+CD8T+ CTLs in the treated tumors. Consistent with those findings, addition of anti-PD1 antibody promoted OVA-specific CTL activation in vitro while addition of PGE2 strongly inhibited it, as measured by IFNγ secretion. Inclusion of E7046 dose-dependently reversed the PGE2-induced suppressive activity in the presence of anti-PD1 antibody. Among major human PGE2 pathway genes, TCGA analysis showed that PTGES1 was upregulated and HPGD downregulated across a broad range of tumor types. In contrast, COX1, COX2 and PGT showed less difference between malignant and normal tissues. Importantly, these differences of one or multiple PGE2 pathway genes were strongly associated with patient survival in certain cancer types. Conclusions: A subset of human cancer types displays upregulated PGE2 pathway that is associated with a poorer prognosis. PGE2-EP4 signaling potently suppresses antigen-specific CTL activation in the presence of PD1 signaling blockade. The combination of EP4 antagonist E7046 with either anti-PD1 or anti-CTLA4 demonstrated superior anti-tumor activity compared with anti-PD1 or anti-CTLA4 alone. This increased activity was accompanied by increased CTL activation. Citation Format: Diana I. Albu, David Verbel, Yuan Huang, Donna Kolber-Simonds, Zichun Wang, Xulong Wang, Zoltan Dezso, Christy Ingersoll, Kuan-Chun Huang, Janna Hutz, Mary Woodall-Jappe, Xingfeng Bao. Specific inhibition of PGE2-EP4 signaling by E7046 promotes anti-tumor activity of checkpoint blockade agents through boosting cytotoxic T cell activity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4607. doi:10.1158/1538-7445.AM2017-4607


Cancer immunology research | 2016

Abstract B034: Preclinical immune antitumor activity of myeloid-targeting E7046 and Treg depleting E7777

Diana I. Albu; Kuan-Chun Huang; Jiayi Wu; Xingfeng Bao; Kenichi Nomoto; Mary Woodall-Jappe

Purpose: To assess the combined activity of E7046 and E7777 against multiple murine syngeneic tumor models. Background: Both regulatory T cells (Tregs)and myeloid cells are significant constituents in the tumor microenvironment in many types of cancer, where they help to maintain an immunosuppressive milieu that inhibits cytotoxic T cell function, which favors tumor persistence. Prostaglandin E2 (PGE2) engages the EP4 receptor on monocytes to direct myeloid cell activities away from antigen presentation and toward immunosuppression (wound healing). PGE2-driven myeloid responses have been reported to enhance Treg immunosuppressive activity, while Tregs with high expression of the FoxP3 transcription factor have been reported to up-regulate cyclooxygenase expression and PGE2 secretion. E7046 is a highly selective small molecule that potently competes with PGE2 binding to EP4. Single agent E7046 slows the growth of multiple syngeneic murine tumor types, in a mechanism dependent upon the presence of T cells. Denileukin diftitox (ONTAK®), an IL-2/diphtheria toxin fusion protein, was originally approved by the US FDA for treating patients with relapsed/refractory cutaneous T cell lymphoma. E7777 is a new version of denileukin diftitox developed using an improved manufacturing process. We and others have shown that denileukin diftitox selectively depletes Tregs. Neither E7046 nor E7777 affects tumor cell viability in vitro. Here we assessed their combined activity against syngeneic tumor models and the mechanisms driving their activity. Methods: Tumors were implanted subcutaneously in groups of 5-10 female BALB/c mice. When tumors were > 50 mm 3 , mice were treated with E7046 (150 mg/kg, QD x 21, p.o.) and/or E7777 (2.5 or 3 μg/head, Q7D x 2 or 3, i.v). Tumors, spleens, and tumor-draining lymph nodes were excised and analyzed by flow cytometry for immune cell composition and function. Results: As single agents, both E7046 and E7777 delayed the growth of established tumors. When combined, their anti-tumor activities were significantly enhanced, with up to 20% of animals rendered stably tumor-free. Activity was significantly reduced if animals were treated with antibody to deplete CD8 + T cells. In the tumors, the combination treatment dramatically increased the ratio of CD8 T cells to CD4 + CD25 + Foxp3 + Tregs, and also decreased the frequency of immunosuppressive myeloid cells. Importantly, the CD8 + T cells in the treated tumors, but not in the control tumors, were found to express granzyme B. Spleens from the treated animals also showed an increased ratio of CD8 + T cells vs the highly active CD4 + CD25 + Foxp3 + ICOS + Tregs. Conclusions: The combination of E7046 and E7777 showed promising preclinical anti-tumor activity via an immune-mediated mechanism. Citation Format: Diana I. Albu, Kuan-Chun Huang, Jiayi Wu, Xingfeng Bao, Kenichi Nomoto, Mary Woodall-Jappe. Preclinical immune antitumor activity of myeloid-targeting E7046 and Treg depleting E7777. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B034.


Cancer Research | 2014

Abstract 2733: Antitumor activity of the PARP inhibitor E7449 in Ewing's sarcoma

Sharon McGonigle; Zhihong Chen; Jingzang Tao Miu; Kuan-Chun Huang; Donna Kolber-Simonds; Nanding Zhao; Natalie C. Twine; Qiongfang Cao; Galina Kuznetsov; Shanqin Xu; Kenichi Nomoto

Ewing9s sarcoma affects mostly adolescents and young adults with tumors that predominate in bone, characterized by the presence of fusion proteins created by chromosomal translocations (EWS-FLI1, EWS-ERG etc.). Recent studies demonstrated an interaction between EWS fusion proteins and PARP1; increased DNA damage and elevated PARP levels upon fusion protein expression as well as sensitivity to PARP inhibition have been reported. E7449, a potent, orally available PARP inhibitor was evaluated in various xenograft models of Ewing9s sarcoma as a single agent and in combination with TMZ or irinotecan, chemotherapies often used in relapsed patients. In an RD-ES (ATCC® HTB166™) Ewing9s sarcoma (EWS-FLI1) s.c. xenograft model, treatment with single agent E7449 or TMZ resulted in no antitumor activity. However, when combined exquisite synergy that resulted in tumor regression and tumor-free mice was observed, even at the low E7449 combination dose of 1 mg/kg. E7449 dose responsive re-growth of tumors was observed and 8/8 mice remained tumor-free in the highest dose group (30 mg/kg) at study termination. Enhanced toxicity as measured by body weight loss was observed in combination treatment groups but mice generally recovered well. Significant antitumor activity was observed for irinotecan alone in the RD-ES model; combination with E7449 enhanced that activity. Combining TMZ and irinotecan was also efficacious in the RD-ES model. Antitumor activity was further enhanced by the addition of E7449 which also potentiated combination toxicity. Studies are ongoing to optimize dosing of E7449 and chemotherapy and to identify a treatment schedule that maximizes activity and tolerability of the various combinations. Additionally, the activity of E7449 alone and in combination was evaluated in 2 Ewing9s sarcoma patient-derived xenograft (PDx) models. In one model (CTG-0143) neither TMZ nor E7449 as single agents had significant antitumor activity. However, the combination of E7449 + TMZ resulted in striking synergy and tumor regression. Although tumors were large when treatment started, significant antitumor activity was also observed with single agent irinotecan and combination treatment. The second model (CTG-0142) was sensitive to E7449 alone at high dose and to both chemotherapies (TMZ and irinotecan) as single agents. Addition of E7449 to either TMZ or irinotecan resulted in enhanced anti-tumor activity as measured by significantly delayed tumor re-growth following regression. Biomarker analysis is ongoing, including gene expression profiling and assessment of the DNA damage response in tumors from RD-ES and PDx models following drug treatments. In conclusion, the combination of E7449 with TMZ and irinotecan resulted in highly potent anti-cancer activity against cell line and PDx models of Ewing9s sarcoma. These data support the assessment of E7449 as a combination therapy for Ewing9s sarcoma in clinical trials. Citation Format: Sharon McGonigle, Zhihong Chen, Jingzang Tao Miu, Kuan-Chun Huang, Donna Kolber-Simonds, Nanding Zhao, Natalie C. Twine, Qiongfang Cao, Galina Kuznetsov, Shanqin Xu, Kenichi Nomoto. Antitumor activity of the PARP inhibitor E7449 in Ewing9s sarcoma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2733. doi:10.1158/1538-7445.AM2014-2733

Collaboration


Dive into the Kuan-Chun Huang's collaboration.

Researchain Logo
Decentralizing Knowledge