Network


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

Hotspot


Dive into the research topics where Patrick Strout is active.

Publication


Featured researches published by Patrick Strout.


Journal of Controlled Release | 2016

Rational design, biophysical and biological characterization of site-specific antibody-tubulysin conjugates with improved stability, efficacy and pharmacokinetics

Pamela Thompson; Ryan Fleming; Binyam Bezabeh; Fengying Huang; Shenlan Mao; Cui Chen; Jay Harper; Haihong Zhong; Xizhe Gao; Xiang-Qing Yu; Mary Jane Hinrichs; Molly Reed; Adeela Kamal; Patrick Strout; Song Cho; Rob Woods; Robert E. Hollingsworth; Rakesh Dixit; Herren Wu; Changshou Gao; Nazzareno Dimasi

Antibody-drug conjugates (ADCs) are among the most promising empowered biologics for cancer treatment. ADCs are commonly prepared by chemical conjugation of small molecule cytotoxic anti-cancer drugs to antibodies through either lysine side chains or cysteine thiols generated by the reduction of interchain disulfide bonds. Both methods yield heterogeneous conjugates with complex biophysical properties and suboptimal serum stability, efficacy, and pharmacokinetics. To limit the complexity of cysteine-based ADCs, we have engineered and characterized in vitro and in vivo antibody cysteine variants that allow precise control of both site of conjugation and drug load per antibody molecule. We demonstrate that the chemically-defined cysteine-engineered antibody-tubulysin conjugates have improved ex vivo and in vivo stability, efficacy, and pharmacokinetics when compared to conventional cysteine-based ADCs with similar drug-to-antibody ratios. In addition, to limit the non-target FcγRs mediated uptake of the ADCs by cells of the innate immune system, which may result in off-target toxicities, the ADCs have been engineered to lack Fc-receptor binding. The strategies described herein are broadly applicable to any full-length IgG or Fc-based ADC and have been incorporated into an ADC that is in phase I clinical development.


Methods of Molecular Biology | 2013

Selecting an Optimal Antibody for Antibody–Drug Conjugate Therapy: Internalization and Intracellular Localization

Jay Harper; Shenlan Mao; Patrick Strout; Adeela Kamal

Antibody-drug conjugates (ADCs) combine the selectivity of a monoclonal antibody with the killing potency of a cytotoxic drug. For an antibody to function as a successful component of an ADC, it needs to bind to the target antigen on the surface of tumor cells and then be internalized by the cell. Following internalization, the ADC has to be transported to the lysosome where subsequent intracellular processing of the ADC will release the biologically active drug to exert its cytotoxic effects on tumor cells. This chapter describes some of the techniques that are currently used to determine internalization and proper intracellular trafficking of antibodies in order to select an optimal antibody for ADC therapeutics.


Bioconjugate Chemistry | 2015

Hydrolytically Stable Site-Specific Conjugation at the N-Terminus of an Engineered Antibody

Pamela Thompson; Binyam Bezabeh; Ryan Fleming; Monica Pruitt; Shenlan Mao; Patrick Strout; Cui Chen; Song Cho; Haihong Zhong; Herren Wu; Changshou Gao; Nazzareno Dimasi

Antibody-drug conjugates (ADCs) have emerged as an important class of therapeutics for cancer treatment that combine the target specificity of antibodies with the killing activity of anticancer chemotherapeutics. Early conjugation technologies relied upon random conjugation to either lysine or cysteine residues, resulting in heterogeneous ADCs. Recent technology advancements have resulted in the preparation of homogeneous ADCs through the site-specific conjugation at engineered cysteines, glycosylated amino acids, and bioorthogonal unnatural amino acids. Here we describe for the first time the conjugation of an anti-mitotic drug to an antibody following the mild and selective oxidation of a serine residue engineered at the N-terminus of the light chain. Using an alkoxyamine-derivatized monomethyl auristatine E payload, we have prepared a hydrolytically stable ADC that retains binding to its antigen and displays potent in vitro cytotoxicity and in vivo tumor growth inhibition.


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.


Molecular Cancer Therapeutics | 2013

Abstract A49: Anti-DLL4 antibodies inhibit cancer stem cells in small cell lung cancer.

Patrick Strout; Martin Korade; Ching Ching Leow; Ivan Inigo; Suneetha Thomas; Elaine M. Hurt; Jon Chesebrough; Adeela Kamal; Song Cho

Small cell lung cancer (SCLC) is a very aggressive lung cancer with features suggesting enrichment in cancer stem cells (CSCs). Delta-like ligand 4 (DLL4) is a membrane bound ligand for Notch receptors critical for functional angiogenesis. Blocking DLL4 signaling increases the density of nonfunctional blood vessels and hypoxia of tumors, and thereby inhibits growth of tumor xenografts in mice. In addition, growing evidence implicates DLL4 Notch signaling pathway in the maintenance of CSCs. Here we investigated the anti-CSC activity of anti-DLL4 mAbs using in vitro and in vivo models of SCLC. Biochemical and flow cytometry analyses revealed that multiple SCLC cell lines express DLL4, and notably, SCLC spheres cultured under CSC-enriching conditions express higher levels of DLL4. Prior to determining the ability of anti-DLL4 mAb in inhibiting CSC in vivo, we first characterized the phenotype of CSCs in the SCLC cell line NCI-H69. NCI-H69 cells enriched for high CD133 expression were more tumorigenic and expressed higher levels of Nanog, Oct 3/4, and EZH2, which are genes crucial for maintenance of CSCs, than cells with low or negative levels of CD133. Moreover, RNA level of DLL4 was found to be three fold higher in CD133 high cells compared to CD133 low cells, suggesting that expression of DLL4 correlates with the CSC phenotype. In vivo, an anti-DLL4 antibody leads to inhibition of NCI-H69 tumor growth when the antibody is administered as a single agent, or in combination with cisplatin+etoposide or topotecan, chemotherapy agents commonly used in treatment of SCLC. To determine if blockade of DLL4 inhibits CSCs in vivo, NCI-H69 tumor xenografts treated with anti-DLL4 mAb were analyzed by flow cytometry. A subset of dissociated tumor cells expressed CD133 and DLL4, and a reduction of these populations of cells was observed in the anti-DLL4 antibody treated groups. In summary, these studies highlight that, in addition to vascular expression, (1) DLL4 is frequently expressed in SCLC cells, (2) DLL4 expression correlates with a CSC phenotype, and (3) that DLL4 blockade using an anti-DLL4 mAb results in inhibition of CSCs. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):A49. Citation Format: Patrick Strout, Martin Korade, Ching Ching Leow, Ivan Inigo, Suneetha Thomas, Elaine Hurt, Jon Chesebrough, Adeela Kamal, Song Cho. Anti-DLL4 antibodies inhibit cancer stem cells in small cell lung cancer. [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 A49.


Molecular Cancer Therapeutics | 2018

Antitumor Activity of MEDI3726 (ADCT-401), a Pyrrolobenzodiazepine Antibody–Drug Conjugate Targeting PSMA, in Preclinical Models of Prostate Cancer

Song Cho; Francesca Zammarchi; David G. Williams; Carin E.G. Havenith; Noel R. Monks; Peter Tyrer; Francois D'Hooge; Ryan Fleming; Kapil Vashisht; Nazzareno Dimasi; Francois Bertelli; Simon Corbett; Lauren Adams; Halla W. Reinert; Sandamali Dissanayake; Charles E. Britten; Wanda King; Karma Dacosta; Ravinder Tammali; Kevin Schifferli; Patrick Strout; Martin Korade; Mary Jane Hinrichs; Simon Chivers; Eva Corey; He Liu; Sae Kim; Neil H. Bander; Philip W. Howard; John A. Hartley

Prostate-specific membrane antigen (PSMA) is a membrane-bound glutamate carboxypeptidase that is highly expressed in nearly all prostate cancers with the highest expression in metastatic castration-resistant prostate cancer (mCRPC). The prevalence of increased surface expression and constitutive internalization of PSMA make it an attractive target for an antibody–drug conjugate (ADC) approach to treating patients with mCRPC. MEDI3726 (previously known as ADCT-401) is an ADC consisting of an engineered version of the anti-PSMA antibody J591 site specifically conjugated to the pyrrolobenzodiazepine (PBD) dimer tesirine. MEDI3726 specifically binds the extracellular domain of PSMA and, once internalized, releases the PBD dimer to crosslink DNA and trigger cell death. In vitro, MEDI3726 demonstrated potent and specific cytotoxicity in a panel of PSMA-positive prostate cancer cell lines, consistent with internalization and DNA interstrand crosslinking. In vivo, MEDI3726 showed robust antitumor activity against the LNCaP and the castration-resistant CWR22Rv1 prostate cancer cell line xenografts. MEDI3726 also demonstrated durable antitumor activity in the PSMA-positive human prostate cancer patient–derived xenograft (PDX) LuCaP models. This activity correlated with increased phosphorylated Histone H2AX in tumor xenografts treated with MEDI3726. MEDI3726 is being evaluated in a phase I clinical trial as a treatment for patients with metastatic castrate-resistant prostate cancer (NCT02991911). Mol Cancer Ther; 17(10); 2176–86. ©2018 AACR.


Cancer Research | 2018

Abstract A004: MEDI3726 (ADCT-401), a novel antibody-drug conjugate targeting PSMA, has potent in vivo antitumor activity in prostate cancer patient-derived xenograft models

Song Cho; Francesca Zammarchi; Noel R. Monks; Kapil Vashisht; Ravinder Tammali; Kevin Schifferli; Patrick Strout; Wanda King; Karma Dacosta; Ryan Fleming; David G. Williams; Karin Havenith; Mary Jane Hinrichs; Simon Chivers; Nazzareno Dimasi; Phil W. Howard; John A. Hartley; Steve Coats; Ronald Herbst; Patrick H. van Berkel; David A. Tice

Prostate specific membrane antigen (PSMA), a type II membrane glycoprotein, is highly expressed in nearly all prostate cancers, with the highest expression in metastatic castration-resistant prostate cancer (mCRPC). The prevalence, increased surface expression in prostate tumors, and constitutive internalization make PSMA an attractive target for an antibody-drug conjugate (ADC) approach in anticancer treatment of patients with mCRPC. MEDI3726 (previously known as ADCT-401) is an ADC consisting of an engineered version of anti-PSMA antibody J591, site-specifically conjugated with DNA cross-linking pyrrolobenzodiazepine (PBD) dimer SG3249 for targeting prostatic cancer cells. Using prostate cancer cell line models, we have previously shown that MEDI3726 specifically binds to the extracellular domain of PSMA and, once internalized, releases the PBD dimer to cross link DNA and achieve potent in vitro and in vivo cytotoxicity. Here we investigated the in vivo activity of MEDI3726 in a series of LuCaP prostate cancer patient-derived xenograft (PDX) models. The selected LuCaP models had varying PSMA expression and heterogeneous genetic and phenotypic backgrounds. In agreement with the earlier cell line xenograft data, dose-dependent antitumor activity was observed in PSMA-positive PDX models with durable tumor regressions in models with high PSMA expression. In the PSMA-negative LuCaP 35CR PDX model, MEDI3726 did not have significant antitumor activity, thus highlighting target-mediated in vivo activity. Increased phosphorylation of histone H2AX was observed in xenografts dosed with MEDI3726, confirming DNA damage induced by interstrand cross-linking PBD dimer as the mechanism of antitumor activity of MEDI3726. In summary, MEDI3726 demonstrated potent and specific in vivo antitumor activity, concurrent with DNA damage, in clinically relevant prostate cancer PDX models. MEDI3726 is being evaluated in phase 1 clinical trial as an anticancer treatment in patients with metastatic castrate-resistant prostate cancer (NCT02991911). Citation Format: Song Cho, Francesca Zammarchi, Noel R. Monks, Kapil Vashisht, Ravinder Tammali, Kevin Schifferli, Patrick Strout, Wanda King, Karma Dacosta, Ryan Fleming, David G. Williams, Karin Havenith, Mary Jane Masson Hinrichs, Simon Chivers, Nazzareno Dimasi, Phil W. Howard, John A. Hartley, Steve Coats, Ronald Herbst, Patrick H. van Berkel, David A. Tice. MEDI3726 (ADCT-401), a novel antibody-drug conjugate targeting PSMA, has potent in vivo antitumor activity in prostate cancer patient-derived xenograft models [abstract]. In: Proceedings of the AACR Special Conference: Prostate Cancer: Advances in Basic, Translational, and Clinical Research; 2017 Dec 2-5; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(16 Suppl):Abstract nr A004.


Transgenic Research | 2017

Generation of a functional humanized Delta-like ligand 4 transgenic mouse model

John Wiseman; Pernilla Gregersson; Johan Johansson; Kerstin Magnell; Fernanda Pilataxi; Chris Morehouse; Philip Brohawn; Nicholas Holoweckyj; Patrick Strout; Song Cho

Humanized mouse models are important tools in many areas of biological drug development including, within oncology research, the development of antagonistic antibodies that have the potential to block tumor growth by controlling vascularization and are key to the generation of in vivo proof-of-concept efficacy data. However, due to cross reactivity between human antibodies and mouse target such studies regularly require mouse models expressing only the human version of the target molecule. Such humanized knock-in/knock-out, KIKO, models are dependent upon the generation of homozygous mice expressing only the human molecule, compensating for loss of the mouse form. However, KIKO strategies can fail to generate homozygous mice, even though the human form is expressed and the endogenous mouse locus is correctly targeted. A typical strategy for generating KIKO mice is by ATG fusion where the human cDNA is inserted downstream of the endogenous mouse promoter elements. However, when adopting this strategy it is possible that the mouse promoter fails to express the human form in a manner compensating for loss of the mouse form or alternatively the human protein is incompatible in the context of the mouse pathway being investigated. So to understand more around the biology of KIKO models, and to overcome our failure with a number of ATG fusion strategies, we developed a range of humanized models focused on Delta-like 4 (Dll4), a target where we initially failed to generate a humanized model. By adopting a broader biologic strategy, we successfully generated a humanized DLL4 KIKO which led to a greater understanding of critical biological aspects for consideration when developing humanized models.


Cancer Research | 2013

Abstract 5098: A mouse DLL4 cross-reactive variant of MEDI0639 disrupts functional vessel formation and inhibits tumor growth in preclinical models.

Song H. Cho; David W. Jenkins; Patrick Strout; Martin Korade; Haihong Zhong; Ching Ching Leow; Shannon Breen; Jon Chesebrough; Nicholas Holoweckyj; Ivan Inigo; Gennady Gololobov; Ping Tsui; Kimberly E. Cook; Melissa Damschroder; Meggan Czapiga; Philip Brohawn; Jiaqi Huang; Sally-Ann Ricketts; Juliana Maynard; Adeela Kamal

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Delta-like ligand 4 (DLL4) is a membrane bound ligand for Notch receptors essential for functional vessel formation during development and angiogenesis. We have shown previously that MEDI0639, a human anti-DLL4 antibody, has potent in vitro and in vivo activity in blocking DLL4-Notch interaction. However, because MEDI0639 has low binding affinity to mouse DLL4, to enable in vivo pharmacology studies using xenograft tumor models in mice, MEDI0639 was engineered to bind more strongly to mouse DLL4. The resulting affinity-optimized variant antibody (anti-mDLL4 mAb) bound with high affinity to both human and mouse DLL4 and maintained binding specificity to DLL4. In a number of human cancer xenograft models in nude mice, the MEDI0639 variant antibody markedly inhibited the growth of tumors. qRT-PCR analysis using the Fluidigm platform showed decreases in mouse genes downstream of the Notch signaling pathway at efficacious doses, indicating on-target antibody effect on tumor vasculature. In addition, IHC and imaging analysis of xenograft tumors from the anti-DLL4 mAb dosed animals showed a dose-dependent increase in microvessel density and decrease in tumor perfusion compared to the control groups. In summary, we describe here the generation of mouse and human DLL4 cross-reactive antibody and the utilization of this antibody in demonstrating a positive correlation between the anticipated mechanism of action of DLL4 blockade on vasculature and inhibition of tumor growth. Citation Format: Song H. Cho, David W. Jenkins, Patrick Strout, Martin Korade, Haihong Zhong, Ching Ching Leow, Shannon Breen, Jon Chesebrough, Nicholas Holoweckyj, Ivan Inigo, Gennady Gololobov, Ping Tsui, Kimberly Cook, Melissa Damschroder, Meggan Czapiga, Philip Brohawn, Jiaqi Huang, Sally-Ann Ricketts, Juliana Maynard, Adeela Kamal. A mouse DLL4 cross-reactive variant of MEDI0639 disrupts functional vessel formation and inhibits 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 5098. doi:10.1158/1538-7445.AM2013-5098

Collaboration


Dive into the Patrick Strout's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge