Andrew G. Polson
Genentech
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Andrew G. Polson.
Blood | 2009
David Dornan; Bennett F; Yung-Hsiang Chen; Mark S. Dennis; Dan L. Eaton; Kristi Elkins; Dorothy French; MaryAnn Go; Andrew Jack; Junutula; Hartmut Koeppen; Jeffrey Lau; Jacqueline McBride; Andy C. Rawstron; Xiaoyan Shi; Nancy Yu; Shang-Fan Yu; Peng Yue; Bing Zheng; Allen Ebens; Andrew G. Polson
Here we describe the generation of an antibody-drug conjugate (ADC) consisting of a humanized anti-CD79b antibody that is conjugated to monomethylauristatin E (MMAE) through engineered cysteines (THIOMABs) by a protease cleavable linker. By using flow cytometry, we detected the surface expression of CD79b in almost all non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia patients, suggesting that anti-CD79b-vcMMAE could be widely used in these malignancies. By using NHL cell lines to simulate a patient population we discovered that a minimal cell-surface expression level of CD79b was required for in vitro activity. Within the subpopulation of cell lines above this minimal threshold, we found that sensitivity to free MMAE, mutation of cancer genes, and cell doubling time were poorly correlated with in vitro activity; however, the expression level of BCL-XL was correlated with reduced sensitivity to anti-CD79b-vcMMAE. This observation was supported by in vivo data showing that a Bcl-2 family inhibitor, ABT-263, strikingly enhanced the activity of anti-CD79b-vcMMAE. Furthermore, anti-CD79b-vcMMAE was significantly more effective than a standard-of-care regimen, R-CHOP (ie, rituximab with a single intravenous injection of 30 mg/kg cyclophosphamide, 2.475 mg/kg doxorubicin, 0.375 mg/kg vincristine, and oral dosing of 0.15 mg/kg prednisone once a day for 5 days), in 3 xenograft models of NHL. Together, these data suggest that anti-CD79b-vcMMAE could be broadly efficacious for the treatment of NHL.
Leukemia | 2010
Andrew G. Polson; Marna Williams; A. M. Gray; Reina N. Fuji; Kirsten Achilles Poon; Jacqueline McBride; Helga Raab; T. Januario; MaryAnn Go; Jeffrey Lau; Shang-Fan Yu; Changchun Du; Franklin Fuh; C. Tan; Y. Wu; W.-C. Liang; Saileta Prabhu; J.-P. Stephan; J.-A. Hongo; Randall Dere; Rong Deng; M. Cullen; R de Tute; Fiona Bennett; Andy C. Rawstron; Andrew Jack; Allen Ebens
Antibody-drug conjugates (ADCs) are potent cytotoxic drugs linked to antibodies through chemical linkers, and allow specific targeting of drugs to neoplastic cells. The expression of CD22 is limited to B-cells, and we show that CD22 is expressed on the vast majority of non-Hodgkins lymphomas (NHLs). An ideal target for an ADC for the treatment of NHL would have limited expression outside the B-cell compartment and be highly effective against NHL. We generated an ADC consisting of a humanized anti-CD22 antibody conjugated to the anti-mitotic agent maytansine with a stable linker (anti-CD22-MCC-DM1). Anti-CD22-MCC-DM1 was broadly effective in in vitro killing assays on NHL B-cell lines. We did not find a strong correlation between in vitro potency and CD22 surface expression, internalization of ADC or sensitivity to free drug. We show that anti-CD22-MCC-DM1 was capable of inducing complete tumor regression in NHL xenograft mouse models. Further, anti-CD22-MCC-DM1 was well tolerated in cynomolgus monkeys and substantially decreased circulating B-cells as well as follicle size and germinal center formation in lymphoid organs. These results suggest that anti-CD22-MCC-DM1 has an efficacy, safety and pharmacodynamic profile that support its use as a treatment for NHL.
Cancer Research | 2014
Teemu T. Junttila; Ji Li; Jennifer Johnston; Maria Hristopoulos; Robyn Clark; Diego Ellerman; Bu-Er Wang; Yijin Li; Mary Mathieu; Guangmin Li; Judy Young; Elizabeth Luis; Gail Lewis Phillips; Eric Stefanich; Christoph Spiess; Andrew G. Polson; Bryan Irving; Justin Scheer; Melissa R. Junttila; Mark S. Dennis; Robert F. Kelley; Klara Totpal; Allen Ebens
Clinical results from the latest strategies for T-cell activation in cancer have fired interest in combination immunotherapies that can fully engage T-cell immunity. In this study, we describe a trastuzumab-based bispecific antibody, HER2-TDB, which targets HER2 and conditionally activates T cells. HER2-TDB specifically killed HER2-expressing cancer cells at low picomolar concentrations. Because of its unique mechanism of action, which is independent of HER2 signaling or chemotherapeutic sensitivity, HER2-TDB eliminated cells refractory to currently approved HER2 therapies. HER2-TDB exhibited potent antitumor activity in four preclinical model systems, including MMTV-huHER2 and huCD3 transgenic mice. PD-L1 expression in tumors limited HER2-TDB activity, but this resistance could be reversed by anti-PD-L1 treatment. Thus, combining HER2-TDB with anti-PD-L1 yielded a combination immunotherapy that enhanced tumor growth inhibition, increasing the rates and durability of therapeutic response.
Molecular Cancer Therapeutics | 2013
Dongwei Li; Kirsten Achilles Poon; Shang-Fan Yu; Randall Dere; MaryAnn Go; Jeffrey Lau; Bing Zheng; Kristi Elkins; Dimitry M. Danilenko; Katherine R. Kozak; Pamela Chan; Josefa Chuh; Xiaoyan Shi; Denise Nazzal; Franklin Fuh; Jacqueline McBride; Vanitha Ramakrishnan; Ruth de Tute; Andy C. Rawstron; Andrew Jack; Rong Deng; Yu-Waye Chu; David Dornan; Marna Williams; William Ho; Allen Ebens; Saileta Prabhu; Andrew G. Polson
Antibody–drug conjugates (ADC), potent cytotoxic drugs linked to antibodies via chemical linkers, allow specific targeting of drugs to neoplastic cells. We have used this technology to develop the ADC DCDT2980S that targets CD22, an antigen with expression limited to B cells and the vast majority of non-Hodgkin lymphomas (NHL). DCDT2980S consists of a humanized anti-CD22 monoclonal IgG1 antibody with a potent microtubule-disrupting agent, monomethyl auristatin E (MMAE), linked to the reduced cysteines of the antibody via a protease cleavable linker, maleimidocaproyl-valine-citrulline-p-aminobenzoyloxycarbonyl (MC-vc-PAB). We describe the efficacy, safety, and pharmacokinetics of DCDT2980S in animal models to assess its potential as a therapeutic for the treatment of B-cell malignancies. We did not find a strong correlation between in vitro or in vivo efficacy and CD22 surface expression, nor a correlation of sensitivity to free drug and in vitro potency. We show that DCDT2980S was capable of inducing complete tumor regression in xenograft mouse models of NHL and can be more effective than rituximab plus combination chemotherapy at drug exposures that were well tolerated in cynomolgus monkeys. These results suggest that DCDT2980S has an efficacy, safety, and pharmacokinetics profile that support potential treatment of NHL. Mol Cancer Ther; 12(7); 1255–65. ©2013 AACR.
Clinical Cancer Research | 2015
Shang-Fan Yu; Bing Zheng; MaryAnn Go; Jeff Lau; Susan D. Spencer; Helga Raab; Robert Soriano; Suchit Jhunjhunwala; Robert L. Cohen; Michele Caruso; Paul Polakis; John A. Flygare; Andrew G. Polson
Purpose: We are interested in identifying mechanisms of resistance to the current generation of antibody–drug conjugates (ADC) and developing ADCs that can overcome this resistance. Experimental Design: Pinatuzumab vedotin (anti-CD22-vc-MMAE) and polatuzumab vedotin (anti-CD79b-vc-MMAE) are ADCs that contain the microtubule inhibitor monomethyl auristatin E (MMAE) attached to the antibody by the protease-cleavable linker maleimidocaproyl-valine-citrulline-p-aminobenzoyloxycarbonyl (MC-vc-PAB). Early clinical trial data suggest that these ADCs have promising efficacy for the treatment of non-Hodgkin lymphoma (NHL); however, some patients do not respond or become resistant to the ADCs. Anthracyclines are very effective in NHL, but ADCs containing the anthracycline doxorubicin were not clinically efficacious probably due to the low drug potency and inadequate linker technology. The anthracycline analogue PNU-159682 is thousands of times more cytotoxic than doxorubicin, so we used it to develop a new class of ADCs. We used the same MC-vc-PAB linker and antibody in pinatuzumab vedotin but replaced the MMAE with a derivative of PNU-159682 to make anti-CD22-NMS249 and tested it for in vivo efficacy in xenograft tumors resistant to MMAE-based ADCs. Results: We derived cell lines from in vivo xenograft tumors that were made resistant to anti-CD22-vc-MMAE and anti-CD79b-vc-MMAE. We identified P-gp (ABCB1/MDR1) as the major driver of resistance to the vc-MMAE–based conjugates. Anti-CD22-NMS249 was at least as effective as anti-CD22-vc-MMAE in xenograft models of the parental cell lines and maintained its efficacy in the resistant cell lines. Conclusions: These studies provide proof of concept for an anthracycline-based ADC that could be used to treat B-cell malignancies that are resistant to vc-MMAE conjugates. Clin Cancer Res; 21(14); 3298–306. ©2015 AACR.
Science Translational Medicine | 2015
Melissa R. Junttila; Weiguang Mao; Xi Wang; Bu-Er Wang; Thinh Pham; John A. Flygare; Shang-Fan Yu; Sharon Yee; David M. Goldenberg; Carter Fields; Jeffrey Eastham-Anderson; Mallika Singh; Rajesh Vij; Jo-Anne Hongo; Ron Firestein; Melissa Schutten; Kelly Flagella; Paul Polakis; Andrew G. Polson
An antibody-drug conjugate targeting LGR5 effectively treats intestinal cancer in preclinical models. Stemming the progression of cancer LGR5 is a well-known marker of intestinal cancer stem cells, which makes it an attractive target for anticancer treatments. Unfortunately, it is also found in healthy intestinal stem cells, giving rise to concerns about the potential toxicity of such treatments. Now, Junttila et al. used preclinical models of intestinal cancer to demonstrate that targeting LGR5 with an antibody-drug conjugate is effective for shrinking tumors without damaging the surrounding normal tissues. These observations of preclinical effectiveness as well as safety suggest that targeting LGR5-expressing cells may be a viable therapeutic strategy and a candidate for evaluation in human studies. Cancer stem cells (CSCs) are hypothesized to actively maintain tumors similarly to how their normal counterparts replenish differentiated cell types within tissues, making them an attractive therapeutic target for the treatment of cancer. Because most CSC markers also label normal tissue stem cells, it is unclear how to selectively target them without compromising normal tissue homeostasis. We evaluated a strategy that targets the cell surface leucine-rich repeat–containing G protein–coupled receptor 5 (LGR5), a well-characterized tissue stem cell and CSC marker, with an antibody conjugated to distinct cytotoxic drugs. One antibody-drug conjugate (ADC) demonstrated potent tumor efficacy and safety in vivo. Furthermore, the ADC decreased tumor size and proliferation, translating to improved survival in a genetically engineered model of intestinal tumorigenesis. These data demonstrate that ADCs can be leveraged to exploit differences between normal and cancer stem cells to successfully target gastrointestinal cancers.
Science Translational Medicine | 2011
Bart Burington; Peng Yue; Xiaoyan Shi; Ranjana H. Advani; Jeffrey Lau; Jenille Tan; Susanna Stinson; Jeremy Stinson; Thomas Januario; Sven de Vos; Stephen M. Ansell; Andres Forero-Torres; Grazyna Fedorowicz; Teddy T C Yang; Kristi Elkins; Changchun Du; Sankar Mohan; Nancy Yu; Zora Modrusan; Somasekar Seshagiri; Shang Fan Yu; Ajay Pandita; Hartmut Koeppen; Dorothy French; Andrew G. Polson; Rienk Offringa; Nancy Whiting; Allen Ebens; David Dornan
A 15-gene expression signature predicts whether a patient with diffuse large B cell lymphoma will respond to dacetuzumab, a therapeutic antibody. Matching Treatment to Tumor If physicians could predict the future, it would take the guess work out of designing the right treatment regimen for every patient’s cancer. The results presented by Burington et al. move us closer to clearing the crystal ball for diffuse large B cell lymphomas, a common type of non-Hodgkin’s lymphoma in which a cell surface receptor, CD40, presents a seemingly attractive target for therapy. Although stimulation of CD40 by ligand binding can cause apoptosis of B cells—a trait that one would predict to be desirable for a B cell lymphoma drug—it can also induce undesirable proliferation of some lymphoma cells. This murky paradox makes it unclear when to prescribe dacetuzumab, a CD40-targeted therapeutic monoclonal antibody. The authors have now identified a 15-gene expression signature that signals the biochemical status of a lymphoma, thus clarifying whether it can be subdued by anti-CD40 therapy. The authors collected an array of cell lines derived from non-Hodgkin’s lymphomas that show a range of sensitivity to anti-CD40 therapy. By assessing gene expression before and after CD40 stimulation and creating a score that reflected CD40 pathway activation, Burington et al. found that cell lines with higher baseline activation of the CD40 pathway tended to be unresponsive to anti-CD40 stimulation. The researchers then identified a group of 15 active genes whose expression in formalin-fixed tissue (as would be obtained from patients) correctly predicted susceptibility to anti-CD40 treatment 77% of the time, a result verified in another set of cells lines and by quantitative polymerase chain reaction (PCR). Next, in a real-world test of the utility of this 15-gene predictor, tumor tissue samples from 39 patients who had been treated with dacetuzumab were scored for CD40 pathway activation with the new gene signature. A large majority (88%) of the patients predicted by the gene signature to be resistant to therapy in fact did not respond to therapy, showing a median progression-free survival of 40 days; 67% of those predicted to respond to dacetuzumab did so, with median progression-free survival extended to 169 days. These results encourage further testing in a prospective clinical trial designed to examine the ability of the 15-gene signature to predict which lymphoma patients will benefit from dacetuzumab treatment. If this index proves useful, it can be added to the catalog of prognostic tools at the service of the oncologist as they match drug to patient—without the need of a crystal ball. The primary function of B cells, critical components of the adaptive immune response, is to produce antibodies against foreign antigens, as well as to perform isotype class switching, which changes the heavy chain of an antibody so that it can interact with different repertoires of effector cells. CD40 is a member of the tumor necrosis factor superfamily of cell surface receptors that transmits survival signals to B cells. In contrast, in B cell cancers, stimulation of CD40 signaling results in a heterogeneous response in which cells can sometimes undergo cell death in response to treatment, depending on the system studied. We found an association between sensitivity to CD40 stimulation and mutation of the tumor suppressor p53 in a panel of non-Hodgkin’s lymphoma cell lines. Consistent with p53’s tumor suppressor role, we found that higher levels of intrinsic DNA damage and increased proliferation rates, as well as higher levels of BCL6, a transcriptional repressor proto-oncogene, were associated with sensitivity to CD40 stimulation. In addition, CD40 treatment–resistant cell lines were sensitized to CD40 stimulation after the introduction of DNA-damaging agents. Using gene expression analysis, we also showed that resistant cell lines exhibited a preexisting activated CD40 pathway and that an mRNA expression signature comprising CD40 target genes predicted sensitivity and resistance to CD40-activating agents in cell lines and mouse xenograft models. Finally, the gene signature predicted tumor shrinkage and progression-free survival in patients with diffuse large B cell lymphoma treated with dacetuzumab, a monoclonal antibody with partial CD40 agonist activity. These data show that CD40 pathway activation status may be useful in predicting the antitumor activity of CD40-stimulating therapeutic drugs.
Molecular Pharmaceutics | 2015
Steven R. Leong; Wei Ching Liang; Yan Wu; Lisa Crocker; Eric Cheng; Deepak Sampath; Rachana Ohri; Helga Raab; Philip E. Hass; Thinh Pham; Ron Firestein; Dongwei Li; Melissa Schutten; Nicola J. Stagg; Annie Ogasawara; Neelima Koppada; Leslie Roth; Simon Williams; Byoung Chul Lee; Cecile Chalouni; Ivan Peng; Jason DeVoss; Jarrod Tremayne; Paul Polakis; Andrew G. Polson
B7-H4 has been implicated in cancers of the female reproductive system and investigated for its possible use as a biomarker for cancer, but there are no preclinical studies to demonstrate that B7-H4 is a molecular target for therapeutic intervention of cancer. We provide evidence that the prevalence and expression levels of B7-H4 are high in different subtypes of breast cancer and that only a few normal tissues express B7-H4 on the cell membrane. These profiles of low normal expression and upregulation in cancer provide an opportunity for the use of antibody-drug conjugates (ADCs), cytotoxic drugs chemically linked to antibodies, for the treatment of B7-H4 positive cancers. We have developed an ADC specific to B7-H4 that uses a linker drug consisting of a potent antimitotic, monomethyl auristatin E (MMAE), linked to engineered cysteines (THIOMAB) via a protease labile linker. We will refer to ADCs that use the THIOMAB format as TDCs to help distinguish the format from standard MC-vc-MMAE ADCs that are conjugated to the interchain disulfide bonds. Anti-B7-H4 (h1D11)-MC-vc-PAB-MMAE (h1D11 TDC) produced durable tumor regression in cell line and patient-derived xenograft models of triple-negative breast cancer. It also binds rat B7-H4 with similar affinity to human and allowed us to test for target dependent toxicity in rats. We found that our anti-B7-H4 TDC has toxicity findings similar to untargeted TDC. Our results validate B7-H4 as an ADC target for breast cancer and support the possible use of this TDC in the treatment of B7-H4(+) breast cancer.
Bioorganic & Medicinal Chemistry Letters | 2015
Vishal Verma; Thomas H. Pillow; Laura DePalatis; Guangmin Li; Gail Lewis Phillips; Andrew G. Polson; Helga Raab; Susan D. Spencer; Bing Zheng
The cryptophycins are a potent class of cytotoxic agents that were evaluated as antibody drug conjugate (ADC) payloads. Free cryptophycin analog 1 displayed cell activity an order of magnitude more potent than approved ADC payloads MMAE and DM1. This potency increase was also reflected in the activity of the cryptophycin ADCs, attached via a either cleavable or non-cleavable linker.
Blood | 2017
Steven R. Leong; Siddharth Sukumaran; Maria Hristopoulos; Klara Totpal; Shannon Stainton; Elizabeth Lu; Alfred Wong; Lucinda Tam; Robert J. Newman; Brian R. Vuillemenot; Diego Ellerman; Chen Gu; Mary Mathieu; Mark S. Dennis; Allen Nguyen; Bing Zheng; Crystal Zhang; Genee Lee; Yu-Waye Chu; Rodney A. Prell; Kedan Lin; Steven T. Laing; Andrew G. Polson
Acute myeloid leukemia (AML) is a major unmet medical need. Most patients have poor long-term survival, and treatment has not significantly changed in 40 years. Recently, bispecific antibodies that redirect the cytotoxic activity of effector T cells by binding to CD3, the signaling component of the T-cell receptor, and a tumor target have shown clinical activity. Notably, blinatumomab is approved to treat relapsed/refractory acute lymphoid leukemia. Here we describe the design, discovery, pharmacologic activity, pharmacokinetics, and safety of a CD3 T cell-dependent bispecific (TDB) full-length human IgG1 therapeutic antibody targeting CLL-1 that could potentially be used in humans to treat AML. CLL-1 is prevalent in AML and, unlike other targets such as CD33 and CD123, is not expressed on hematopoietic stem cells providing potential hematopoietic recovery. We selected a high-affinity monkey cross-reactive anti-CLL-1 arm and tested several anti-CD3 arms that varied in affinity, and determined that the high-affinity CD3 arms were up to 100-fold more potent in vitro. However, in mouse models, the efficacy differences were less pronounced, probably because of prolonged exposure to TDB found with lower-affinity CD3 TDBs. In monkeys, assessment of safety and target cell depletion by the high- and low-affinity TDBs revealed that only the low-affinity CD3/CLL1 TDB was well tolerated and able to deplete target cells. Our data suggest that an appropriately engineered CLL-1 TDB could be effective in the treatment of AML.