Network


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

Hotspot


Dive into the research topics where Leslie Wetzel is active.

Publication


Featured researches published by Leslie Wetzel.


Cancer Cell | 2016

A Biparatopic HER2-Targeting Antibody-Drug Conjugate Induces Tumor Regression in Primary Models Refractory to or Ineligible for HER2-Targeted Therapy.

John Y. Li; Samuel R. Perry; Vanessa Muniz-Medina; Xinzhong Wang; Leslie Wetzel; Marlon Rebelatto; Mary Jane Hinrichs; Binyam Bezabeh; Ryan Fleming; Nazzareno Dimasi; Hui Feng; Dorin Toader; Andy Q. Yuan; Lan Xu; Jia Lin; Changshou Gao; Herren Wu; Rakesh Dixit; Jane K. Osbourn; Steven Coats

Antibody-drug conjugate (ADC) which delivers cytotoxic drugs specifically into targeted cells through internalization and lysosomal trafficking has emerged as an effective cancer therapy. We show that a bivalent biparatopic antibody targeting two non-overlapping epitopes on HER2 can induce HER2 receptor clustering, which in turn promotes robust internalization, lysosomal trafficking, and degradation. When conjugated with a tubulysin-based microtubule inhibitor, the biparatopic ADC demonstrates superior anti-tumor activity over ado-trastuzumab emtansine (T-DM1) in tumor models representing various patient subpopulations, including T-DM1 eligible, T-DM1 ineligible, and T-DM1 relapsed/refractory. Our findings indicate that this biparatopic ADC has promising potential as an effective therapy for metastatic breast cancer and a broader patient population may benefit from this unique HER2-targeting ADC.


Neoplasia | 2015

Doxil Synergizes with Cancer Immunotherapies to Enhance Antitumor Responses in Syngeneic Mouse Models

Jonathan Rios-Doria; Nicholas M. Durham; Leslie Wetzel; Raymond Rothstein; Jon Chesebrough; Nicholas Holoweckyj; Wei Zhao; Ching Ching Leow; Robert E. Hollingsworth

Based on the previously described roles of doxorubicin in immunogenic cell death, both doxorubicin and liposomal doxorubicin (Doxil) were evaluated for their ability to boost the antitumor response of different cancer immunotherapies including checkpoint blockers (anti–PD-L1, PD-1, and CTLA-4 mAbs) and TNF receptor agonists (OX40 and GITR ligand fusion proteins) in syngeneic mouse models. In a preventative CT26 mouse tumor model, both doxorubicin and Doxil synergized with anti–PD-1 and CTLA-4 mAbs. Doxil was active when CT26 tumors were grown in immunocompetent mice but not immunocompromised mice, demonstrating that Doxil activity is increased in the presence of a functional immune system. Using established tumors and maximally efficacious doses of Doxil and cancer immunotherapies in either CT26 or MCA205 tumor models, combination groups produced strong synergistic antitumor effects, a larger percentage of complete responders, and increased survival. In vivo pharmacodynamic studies showed that Doxil treatment decreased the percentage of tumor-infiltrating regulatory T cells and, in combination with anti–PD-L1, increased the percentage of tumor-infiltrating CD8+ T cells. In the tumor, Doxil administration increased CD80 expression on mature dendritic cells. CD80 expression was also increased on both monocytic and granulocytic myeloid cells, suggesting that Doxil may induce these tumor-infiltrating cells to elicit a costimulatory phenotype capable of activating an antitumor T-cell response. These results uncover a novel role for Doxil in immunomodulation and support the use of Doxil in combination with checkpoint blockade or TNFR agonists to increase response rates and antitumor activity.


Molecular Cancer Therapeutics | 2016

A Potent HER3 Monoclonal Antibody That Blocks Both Ligand-Dependent and -Independent Activities: Differential Impacts of PTEN Status on Tumor Response

Zhan Xiao; Rosa A. Carrasco; Kevin Schifferli; Krista Kinneer; Ravinder Tammali; Hong Chen; Ray Rothstein; Leslie Wetzel; Chunning Yang; Partha S. Chowdhury; Ping Tsui; Philipp Steiner; Bahija Jallal; Ronald Herbst; Robert E. Hollingsworth; David A. Tice

HER3/ERBB3 is a kinase-deficient member of the EGFR family receptor tyrosine kinases (RTK) that is broadly expressed and activated in human cancers. HER3 is a compelling cancer target due to its important role in activation of the oncogenic PI3K/AKT pathway. It has also been demonstrated to confer tumor resistance to a variety of cancer therapies, especially targeted drugs against EGFR and HER2. HER3 can be activated by its ligand (heregulin/HRG), which induces HER3 heterodimerization with EGFR, HER2, or other RTKs. Alternatively, HER3 can be activated in a ligand-independent manner through heterodimerization with HER2 in HER2-amplified cells. We developed a fully human mAb against HER3 (KTN3379) that efficiently suppressed HER3 activity in both ligand-dependent and independent settings. Correspondingly, KTN3379 inhibited tumor growth in divergent tumor models driven by either ligand-dependent or independent mechanisms in vitro and in vivo. Most intriguingly, while investigating the mechanistic underpinnings of tumor response to KTN3379, we discovered an interesting dichotomy in that PTEN loss, a frequently occurring oncogenic lesion in a broad range of cancer types, substantially blunted the tumor response in HER2-amplified cancer, but not in the ligand-driven cancer. To our knowledge, this represents the first study ascertaining the impact of PTEN loss on the antitumor efficacy of a HER3 mAb. KTN3379 is currently undergoing a phase Ib clinical trial in patients with advanced solid tumors. Our current study may help us optimize patient selection schemes for KTN3379 to maximize its clinical benefits. Mol Cancer Ther; 15(4); 689–701. ©2016 AACR.


Cancer Research | 2017

Antibody-drug conjugates bearing pyrrolobenzodiazepine or tubulysin payloads are immunomodulatory and synergize with multiple immunotherapies

Jonathan Rios-Doria; Jay Harper; Raymond Rothstein; Leslie Wetzel; Jon Chesebrough; Allison M. Marrero; Cui Chen; Patrick Strout; Kathy Mulgrew; Kelly McGlinchey; Ryan Fleming; Binyam Bezabeh; John Meekin; David B. Stewart; Maureen Kennedy; Philip Martin; Andrew Buchanan; Nazzareno Dimasi; Emil Michelotti; Robert E. Hollingsworth

Immunogenic cell death (ICD) is the process by which certain cytotoxic drugs induce apoptosis of tumor cells in a manner that stimulates the immune system. In this study, we investigated whether antibody-drug conjugates (ADCS) conjugated with pyrrolobenzodiazepine dimer (PBD) or tubulysin payloads induce ICD, modulate the immune microenvironment, and could combine with immuno-oncology drugs to enhance antitumor activity. We show that these payloads on their own induced an immune response that prevented the growth of tumors following subsequent tumor cell challenge. ADCs had greater antitumor activity in immunocompetent versus immunodeficient mice, demonstrating a contribution of the immune system to the antitumor activity of these ADCs. ADCs also induced immunologic memory. In the CT26 model, depletion of CD8+ T cells abrogated the activity of ADCs when used alone or in combination with a PD-L1 antibody, confirming a role for T cells in antitumor activity. Combinations of ADCs with immuno-oncology drugs, including PD-1 or PD-L1 antibodies, OX40 ligand, or GITR ligand fusion proteins, produced synergistic antitumor responses. Importantly, synergy was observed in some cases with suboptimal doses of ADCs, potentially providing an approach to achieve potent antitumor responses while minimizing ADC-induced toxicity. Immunophenotyping studies in different tumor models revealed broad immunomodulation of lymphoid and myeloid cells by ADC and ADC/immuno-oncology combinations. These results suggest that it may be possible to develop novel combinatorial therapies with PBD- and tubulysin-based ADC and immuno-oncology drugs that may increase clinical responses. Cancer Res; 77(10); 2686-98. ©2017 AACR.


Molecular Cancer Therapeutics | 2017

Preclinical Evaluation of MEDI0641, a Pyrrolobenzodiazepine-Conjugated Antibody–Drug Conjugate Targeting 5T4

Jay Harper; Chris Lloyd; Nazzareno Dimasi; Dorin Toader; Rose Marwood; Leeanne Lewis; David Bannister; Jelena Jovanović; Ryan Fleming; Francois D'Hooge; Shenlan Mao; Allison M. Marrero; Martin Korade; Patrick Strout; Linda Xu; Cui Chen; Leslie Wetzel; Shannon Breen; Lilian van Vlerken-Ysla; Sanjoo Jalla; Marlon Rebelatto; Haihong Zhong; Elaine M. Hurt; Mary Jane Hinrichs; Keven Huang; Philip W. Howard; David A. Tice; Robert E. Hollingsworth; Ronald Herbst; Adeela Kamal

Antibody–drug conjugates (ADC) are used to selectively deliver cytotoxic agents to tumors and have the potential for increased clinical benefit to cancer patients. 5T4 is an oncofetal antigen overexpressed on the cell surface in many carcinomas on both bulk tumor cells as well as cancer stem cells (CSC), has very limited normal tissue expression, and can internalize when bound by an antibody. An anti-5T4 antibody was identified and optimized for efficient binding and internalization in a target-specific manner, and engineered cysteines were incorporated into the molecule for site-specific conjugation. ADCs targeting 5T4 were constructed by site-specifically conjugating the antibody with payloads that possess different mechanisms of action, either a DNA cross-linking pyrrolobenzodiazepine (PBD) dimer or a microtubule-destabilizing tubulysin, so that each ADC had a drug:antibody ratio of 2. The resulting ADCs demonstrated significant target-dependent activity in vitro and in vivo; however, the ADC conjugated with a PBD payload (5T4-PBD) elicited more durable antitumor responses in vivo than the tubulysin conjugate in xenograft models. Likewise, the 5T4-PBD more potently inhibited the growth of 5T4-positive CSCs in vivo, which likely contributed to its superior antitumor activity. Given that the 5T4-PBD possessed both potent antitumor activity as well as anti-CSC activity, and thus could potentially target bulk tumor cells and CSCs in target-positive indications, it was further evaluated in non-GLP rat toxicology studies that demonstrated excellent in vivo stability with an acceptable safety profile. Taken together, these preclinical data support further development of 5T4-PBD, also known as MEDI0641, against 5T4+ cancer indications. Mol Cancer Ther; 16(8); 1576–87. ©2017 AACR.


Journal of Controlled Release | 2016

Lipid- and polyion complex-based micelles as agonist platforms for TNFR superfamily receptors.

Ryan N. Gilbreth; Shabazz Novarra; Leslie Wetzel; Stelios Florinas; Horacio Cabral; Kazunori Kataoka; Jonathan Rios-Doria; Ronald J. Christie; Manuel Baca

Receptor clustering is important for signaling among the therapeutically relevant TNFR superfamily of receptors. In nature, this clustering is driven by trimeric ligands often presented in large numbers as cell surface proteins. Molecules capable of driving similar levels of clustering could make good agonists and hold therapeutic value. However, recapitulating such extensive clustering using typical biotherapeutic formats, such as antibodies, is difficult. Consequently, generating effective agonists of TNFR superfamily receptors is challenging. Toward addressing this challenge we have used lipid- and polyion complex-based micelles as platforms for presenting receptor-binding biologics in a multivalent format that facilitates receptor clustering and imparts strong agonist activity. We show that receptor-binding scFvs or small antibody mimetics that have no agonist activity on their own can be transformed into potent agonists through multivalent presentation on a micelle surface and that the activity of already active multivalent agonists can be enhanced. Using this strategy, we generated potent agonists against two different TNFR superfamily receptors and mouse tumor model studies demonstrate that these micellar agonists have therapeutic efficacy in vivo. Due to its ease of implementation and applicability independent of agonist molecular format, we anticipate that this strategy could be useful for developing agonists to a variety of receptors that rely on clustering to signal.


Cancer Research | 2016

Abstract 2970: MEDI4276, a HER2-targeting antibody tubulysin conjugate, displays potent in vitro and in vivo activity in preclinical studies

John Y. Li; Dorin Toader; Samuel R. Perry; Vanessa Muniz-Medina; Leslie Wetzel; Marlon Rebelatto; Mary Jane Hinrichs; Ryan Fleming; Binyam Bezabeh; Pamela Thompson; Nazzareno Dimasi; Brandon Lam; Xian-Qing Yu; Changshou Gao; Rakesh Dixit; Steven Coats; Jane K. Osbourn; Herren Wu

Antibody drug conjugates (ADCs) combine the specificity of antibodies with the potent cytotoxicity of small molecule drugs and have shown to provide therapeutic options for various cancers. We report herein the discovery of a HER2-targeting ADC MEDI4276 that showed potent cell killing activity in vitro in cancer cell lines that express the HER2 receptor. The observed in vitro activity translated into in vivo tumor growth inhibition in various xenograft mouse models. MEDI4276 is a homogeneous molecule with precise control of drug loading following site specific conjugation of a cytotoxic drug. The drug in MEDI4276 is MMETA, a fully synthetic analog of the tubulysin family that showed pM potency in a panel of cancer cell lines. MMETA was conjugated to the antibody via engineered cysteines with a maleimide-bearing mc-Lys protease cleavable linker. The antibody in MEDI4276 is a bivalent biparatopic antibody targeting two distinct non-overlapping epitopes on HER2 that leads to antibody-receptor clustering following binding and thus promoting internalization, lysosomal trafficking and degradation. The combination of enhanced internalization and potent cytotoxic drug allows for this ADC to kill tumor cell populations with a broader range of HER2 expression. Preclinical studies showed that MEDI4276 induced tumor regression in HER2-positive tumor models that had developed acquired resistance to T-DM1 and in a number of models with lower HER2 expression that are refractory to T-DM1 treatment. Overall, our findings underscore the potential application of MEDI4276 to treat a large patient population that is ineligible for or relapsed/refractory to current HER2-targeted therapies. MEDI4276 is currently being investigated in a Phase I clinical trial. Citation Format: John Li, Dorin Toader, Samuel R. Perry, Vanessa Muniz-Medina, Leslie Wetzel, Marlon C. Rebelatto, Mary Jane Masson Hinrichs, Ryan Fleming, Binyam Bezabeh, Pamela Thompson, Nazzareno Dimasi, Brandon Lam, Xian-Qing Yu, Changshou Gao, Rakesh Dixit, Steven Coats, Jane Osbourn, Herren Wu. MEDI4276, a HER2-targeting antibody tubulysin conjugate, displays potent in vitro and in vivo activity in preclinical studies. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2970.


Cancer Research | 2013

Abstract 5462: MEDI3185, a potent anti-CXCR4 antibody, inhibits tumor cell migration, signaling and tumor growth in preclinical models.

Adeela Kamal; Youzhen Wang; Philipp Steiner; Anne-Marie Mazzola; Leslie Wetzel; Melissa Passino; Brenda McDermott; Keven Huang; Vahe Bedian; Norman M. Greenberg

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC The chemokine receptor CXCR4 is a seven-transmembrane G-protein coupled receptor that mediates chemotaxis and cell migration upon stimulation via its ligand, stromal-derived factor 1 (SDF-1), also called CXCL12. CXCR4 is normally expressed on bone marrow stem and progenitor cells, various circulating lymphocytes, endothelial precursor cells, tissue macrophages and fibroblasts but the aberrant overexpression of CXCR4 is linked to various hematological malignancies, solid tumors and metastatic neoplasms. Moreover, CXCR4 overexpression is correlated with poor prognosis in many types of cancer, including breast, ovarian, colon, pancreatic, AML and glioblastomas. CXCR4 inhibition using siRNA, small-molecule and peptide inhibitors has demonstrated that it can inhibit tumor growth by blocking tumor cell survival/proliferation, metastasis, angiogenesis and tumor immune infiltrates. Here we describe a novel, fully human, antagonistic antibody to CXCR4, MEDI3185, which blocks SDF-1 binding to CXCR4. MEDI3185 has picomolar binding affinity to human CXCR4 and exhibits no significant binding to other chemokine receptors such as CCR4 or CXCR3. In vitro studies demonstrated that MEDI3185 inhibited tumor cell migration, blocked SDF-1 induced tumor cell signaling and induced apoptosis of tumor cells. In preclinical human tumor xenograft models in mouse, MEDI3185 showed single-agent tumor growth inhibition in multiple myeloma and B-cell Burkitts lymphoma models and had combination activity in ovarian models. In addition, MEDI3185 extended survival as combination therapy in mouse models of CLL and also blocked lung tumor burden in a disseminated ovarian model. Combined, these data suggest that MEDI3185 is a potent CXCR4 antibody for the treatment of both hematological and solid tumors because it has pleiotropic effects on tumor biology that may enhance the efficacy of the current standard of care. Citation Format: Adeela Kamal, Youzhen Wang, Philipp Steiner, Anne-Marie Mazzola, Leslie Wetzel, Melissa Passino, Brenda McDermott, Keven Huang, Vahe Bedian, Norman Greenberg. MEDI3185, a potent anti-CXCR4 antibody, inhibits tumor cell migration, signaling and tumor growth in preclinical models. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5462. doi:10.1158/1538-7445.AM2013-5462


Leukemia | 2018

Preclinical assessment of an antibody–PBD conjugate that targets BCMA on multiple myeloma and myeloma progenitor cells

Krista Kinneer; Matt Flynn; Suneetha Thomas; John Meekin; Reena Varkey; Xiaodong Xiao; Haihong Zhong; Shannon Breen; Paul G. Hynes; Ryan Fleming; Binyam Bezabeh; Cui Chen; Leslie Wetzel; Ruoyan Chen; Nazzareno Dimasi; Yu-Tzu Tai; Kenneth C. Anderson; Ronald Herbst; Philip W. Howard; Elaine M. Hurt; David A. Tice

MP, Ries RE, et al. Expression and functional characterization of CD33 transcript variants in human acute myeloid leukemia. Oncotarget. 2016;7:43281–94. 12. Humbert O, Peterson CW, Norgaard ZK, Radtke S, Kiem HP. A nonhuman primate transplantation model to evaluate hematopoietic stem cell gene editing strategies for betahemoglobinopathies. Mol Ther Methods Clin Dev. 2018;8:75–86. 13. Correnti CE, Laszlo GS, de van der Schueren WJ, Godwin CD, Bandaranayake A, Busch MA, et al. Simultaneous multiple interaction T-cell engaging (SMITE) bispecific antibodies overcome bispecific T-cell engager (BiTE) resistance via CD28 costimulation. Leukemia. 2018;32:1239–43. 14. Haworth KG, Ironside C, Norgaard ZK, Obenza WM, Adair JE, Kiem HP. In vivo murine-matured human CD3(+ ) cells as a preclinical model for T cell-based immunotherapies. Mol Ther Methods Clin Dev. 2017;6:17–30. 15. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671–5.


Molecular Cancer Therapeutics | 2013

Abstract A112: Combined targeting of HER2 and HER3 inhibits tumor growth in both trastuzumab-sensitive and trastuzumab-resistant breast cancer models.

Philipp Steiner; Leslie Wetzel; Kevin Schifferli; Raymond Rothstein; Ravinder Tammali; Marlon Rebelatto; Zhan Xiao; Andrew Pierce; Robert E. Hollingsworth

HER3 (ERBB3) is a tumor driver in divergent cancer types with high HRG levels or heightened EGFR or HER2 kinase activity via growth signal coupling to the PI3K-AKT pathway. HER3 lacks intrinsic tyrosine kinase activity but is capable of signaling after heterodimerizing with tyrosine kinase ERBB family members EGFR or HER2. Heterodimer formation is driven either by the binding of heregulin (NRG1/HRG) to HER3 (ligand dependent - LD), or alternatively, by overexpression of EGFR or HER2 (ligand-independent - LI). Preclinical evaluation of MEDI3379, a human IgG1 modified monoclonal anti-HER3 antibody that antagonizes both LD and LI signaling, demonstrated tumor growth inhibition in the HER2-expressing MDA-MB-361 breast cancer xenograft model. Anti-tumor activity of MEDI3379 in this model was increased in combination with the HER2-targeting drug trastuzumab to a greater extent than MEDI3379 when combined with either lapatinib or pertuzumab. Combined inhibition of HER2 and HER3 led to strong reduction in pHER3. Unexpectedly, co-administration of MEDI3379 together with trastuzumab in MDA-MB-361 tumor-bearing mice but not in naive mice resulted in reduced serum levels of trastuzumab. A control antibody combined with trastuzumab in MDA-MB-361 tumor bearing mice did not have this effect. Furthermore, in the trastuzumab-resistant JIMT-1 breast cancer xenograft model which has high HRG and pHER3 levels MEDI3379 but not pertuzumab resensitized JIMT-1 tumors to trastuzumab resulting in synergistic anti-tumor activity and a reduction of pHER3. In vivo resistance to this regimen appeared with prolonged treatment of JIMT-1 tumors and was accompanied by a decrease in homogenous expression of HER2. In conclusion, the use of MEDI3379 in combination with trastuzumab in HER2-amplifed breast cancer cell lines has demonstrated additional tumor growth inhibition over single agent trastuzumab and overcoming trastuzumab-resistance mechanisms. This combination warrants further evaluation in preclinical and clinical studies. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):A112. Citation Format: Philipp Steiner, Leslie Wetzel, Kevin Schifferli, Raymond Rothstein, Ravinder Tammali, Marlon Rebelatto, Zhan Xiao, Andrew Pierce, Robert Hollingsworth. Combined targeting of HER2 and HER3 inhibits tumor growth in both trastuzumab-sensitive and trastuzumab-resistant breast cancer models. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr A112.

Collaboration


Dive into the Leslie Wetzel's collaboration.

Researchain Logo
Decentralizing Knowledge