34 results on '"Dipti Thakkar"'
Search Results
2. Rationally targeted anti-VISTA antibody that blockades the C-C’ loop region can reverse VISTA immune suppression and remodel the immune microenvironment to potently inhibit tumor growth in an Fc independent manner
- Author
-
Dipti Thakkar, Lillian Liu, Bhushan Dharmadhikari, Shalini Paliwal, Siyu Guan, Shreya Kar, Nikhil K Tulsian, Joshua J Gruber, Leah DiMascio, Konrad H Paszkiewicz, Piers J Ingram, and Jerome D Boyd-Kirkup
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Despite significant progress in cancer immunotherapy in recent years, resistance to existing immune checkpoint therapies (ICT) is common. V-domain Ig suppressor of T cell activation (VISTA), a predominantly myeloid immune checkpoint regulator, represents a promising therapeutic target due to its role in suppressing proinflammatory antitumor responses in myeloid-enriched tumor microenvironments. However, uncertainty around the cognate VISTA ligand has made the development of effective anti-VISTA antibodies challenging. The expression of VISTA on normal immune cell subtypes argues for a neutralizing non-depleting antibody, however, previous reported anti-VISTA antibodies use IgG1 Fc isotypes that deplete VISTA+ cells by antibody dependent cellular cytotoxicity/complement dependent cytotoxicity and these antibodies have shown fast serum clearance and immune toxicities.Method Here we used a rational antibody discovery approach to develop the first Fc-independent anti-VISTA antibody, HMBD-002, that binds a computationally predicted functional epitope within the C-C-loop, distinct from other known anti-VISTA antibodies. This epitope is species-conserved allowing robust in vitro and in vivo testing of HMBD-002 in human and murine models of immune activation and cancer including humanized mouse models.Results We demonstrate here that blockade by HMBD-002 inhibits VISTA binding to potential partners, including V-Set and Immunoglobulin domain containing 3, to reduce myeloid-derived suppression of T cell activity and prevent neutrophil migration. Analysis of immune cell milieu suggests that HMBD-002 treatment stimulates a proinflammatory phenotype characterized by a Th1/Th17 response, recapitulating a phenotype previously noted in VISTA knockout models. This mechanism of action is further supported by immune-competent syngenic and humanized mouse models of colorectal, breast and lung cancer where neutralizing VISTA, without depleting VISTA expressing cells, significantly inhibited tumor growth while decreasing infiltration of suppressive myeloid cells and increasing T cell activity. Finally, we did not observe either the fast serum clearance or immune toxicities that have been reported for IgG1 antibodies.Conclusion In conclusion, we have shown that VISTA-induced immune suppression can be reversed by blockade of the functional C-C’ loop region of VISTA with a first-in-class rationally targeted and non-depleting IgG4 isotype anti-VISTA antibody, HMBD-002. This antibody represents a highly promising novel therapy in the VISTA-suppressed ICT non-responder population.
- Published
- 2022
- Full Text
- View/download PDF
3. Considerations for treatment duration in responders to immune checkpoint inhibitors
- Author
-
Daniel Olson, Jiajia Zhang, Todd Bartkowiak, Abdul Rafeh Naqash, Vaia Florou, Rosa Nguyen, Rania H Younis, Sarah Church, Maria E Rodriguez-Ruiz, Rachel Howard, Thomas U Marron, Ravi B Patel, Michal Sheffer, Abigail Overacre-Delgoffe, Aideen E Ryan, Sangeetha M Reddy, Sabina Kaczanowska, Dipti Thakkar, Kristin G Anderson, Esha Sachdev, Christopher Fuhrman, Jessica E Thaxton, David H Aggen, and Jennifer L Guerriero
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Immune checkpoint inhibitors (ICIs) have improved overall survival for cancer patients, however, optimal duration of ICI therapy has yet to be defined. Given ICIs were first used to treat patients with metastatic melanoma, a condition that at the time was incurable, little attention was initially paid to how much therapy would be needed for a durable response. As the early immunotherapy trials have matured past 10 years, a significant per cent of patients have demonstrated durable responses; it is now time to determine whether patients have been overtreated, and if durable remissions can still be achieved with less therapy, limiting the physical and financial toxicity associated with years of treatment. Well-designed trials are needed to identify optimal duration of therapy, and to define biomarkers to predict who would benefit from shorter courses of immunotherapy. Here, we outline key questions related to health, financial and societal toxicities of over treating with ICI and present four unique clinical trials aimed at exposing criteria for early cessation of ICI. Taken together, there is a serious liability to overtreating patients with ICI and future work is warranted to determine when it is safe to stop ICI.
- Published
- 2021
- Full Text
- View/download PDF
4. The Fc-mediated effector functions of a potent SARS-CoV-2 neutralizing antibody, SC31, isolated from an early convalescent COVID-19 patient, are essential for the optimal therapeutic efficacy of the antibody.
- Author
-
Conrad E Z Chan, Shirley G K Seah, De Hoe Chye, Shane Massey, Maricela Torres, Angeline P C Lim, Steven K K Wong, Jacklyn J Y Neo, Pui San Wong, Jie Hui Lim, Gary S L Loh, Dongling Wang, Jerome D Boyd-Kirkup, Siyu Guan, Dipti Thakkar, Guo Hui Teo, Kiren Purushotorman, Paul E Hutchinson, Barnaby E Young, Jenny G Low, Paul A MacAry, Hannes Hentze, Venkateshan S Prativadibhayankara, Kantharaj Ethirajulu, Jason E Comer, Chien-Te K Tseng, Alan D T Barrett, Piers J Ingram, Trevor Brasel, and Brendon John Hanson
- Subjects
Medicine ,Science - Abstract
Although SARS-CoV-2-neutralizing antibodies are promising therapeutics against COVID-19, little is known about their mechanism(s) of action or effective dosing windows. We report the generation and development of SC31, a potent SARS-CoV-2 neutralizing antibody, isolated from a convalescent patient. Antibody-mediated neutralization occurs via an epitope within the receptor-binding domain of the SARS-CoV-2 Spike protein. SC31 exhibited potent anti-SARS-CoV-2 activities in multiple animal models. In SARS-CoV-2 infected K18-human ACE2 transgenic mice, treatment with SC31 greatly reduced viral loads and attenuated pro-inflammatory responses linked to the severity of COVID-19. Importantly, a comparison of the efficacies of SC31 and its Fc-null LALA variant revealed that the optimal therapeutic efficacy of SC31 requires Fc-mediated effector functions that promote IFNγ-driven anti-viral immune responses, in addition to its neutralization ability. A dose-dependent efficacy of SC31 was observed down to 5mg/kg when administered before viral-induced lung inflammatory responses. In addition, antibody-dependent enhancement was not observed even when infected mice were treated with SC31 at sub-therapeutic doses. In SARS-CoV-2-infected hamsters, SC31 treatment significantly prevented weight loss, reduced viral loads, and attenuated the histopathology of the lungs. In rhesus macaques, the therapeutic potential of SC31 was evidenced through the reduction of viral loads in both upper and lower respiratory tracts to undetectable levels. Together, the results of our preclinical studies demonstrated the therapeutic efficacy of SC31 in three different models and its potential as a COVID-19 therapeutic candidate.
- Published
- 2021
- Full Text
- View/download PDF
5. Supplementary Figure 5 from 10D1F, an Anti-HER3 Antibody That Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Jerome D. Boyd-Kirkup, Piers J. Ingram, Konrad H. Paszkiewicz, Eric Ng, Zhihao Wu, Siyu Guan, Marvin M. Taguiam, Vicente Sancenon, and Dipti Thakkar
- Abstract
10D1P demonstrates potent in vivo tumor growth inhibition in multiple xenograft tumor models
- Published
- 2023
6. Supplementary Figure 4 from 10D1F, an Anti-HER3 Antibody That Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Jerome D. Boyd-Kirkup, Piers J. Ingram, Konrad H. Paszkiewicz, Eric Ng, Zhihao Wu, Siyu Guan, Marvin M. Taguiam, Vicente Sancenon, and Dipti Thakkar
- Abstract
10D1F does not show rapid internalization in HER3 expressing cancer cell lines but activates ADCC
- Published
- 2023
7. Supplementary Material from 10D1F, an Anti-HER3 Antibody That Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Jerome D. Boyd-Kirkup, Piers J. Ingram, Konrad H. Paszkiewicz, Eric Ng, Zhihao Wu, Siyu Guan, Marvin M. Taguiam, Vicente Sancenon, and Dipti Thakkar
- Abstract
Supplementary Methods with References, Supplementary Figure Legends, and list of Supplementary Tables
- Published
- 2023
8. Supplemental Table 2 from 10D1F, an Anti-HER3 Antibody That Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Jerome D. Boyd-Kirkup, Piers J. Ingram, Konrad H. Paszkiewicz, Eric Ng, Zhihao Wu, Siyu Guan, Marvin M. Taguiam, Vicente Sancenon, and Dipti Thakkar
- Abstract
Supplemental Table 2 - Summary of protein and gene expression data of HER family molecules, and their ligands, for the cancer cell lines used in this study
- Published
- 2023
9. Supplementary Figure 3 from 10D1F, an Anti-HER3 Antibody That Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Jerome D. Boyd-Kirkup, Piers J. Ingram, Konrad H. Paszkiewicz, Eric Ng, Zhihao Wu, Siyu Guan, Marvin M. Taguiam, Vicente Sancenon, and Dipti Thakkar
- Abstract
10D1F demonstrates superior inhibition of cancer cell line proliferation by potently inhibiting downstream signaling through the PI3K pathway in additional cancer cell lines
- Published
- 2023
10. Supplementary Figure 1 from 10D1F, an Anti-HER3 Antibody That Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Jerome D. Boyd-Kirkup, Piers J. Ingram, Konrad H. Paszkiewicz, Eric Ng, Zhihao Wu, Siyu Guan, Marvin M. Taguiam, Vicente Sancenon, and Dipti Thakkar
- Abstract
10D1F parental antibody, 10D1P, binds with high affinity and specificity to a species-conserved epitope on HER3, and Class 1 and Class 2 HER3 antibodies show different HER3 binding profiles in presence and absence of NRG1
- Published
- 2023
11. Data from 10D1F, an Anti-HER3 Antibody That Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Jerome D. Boyd-Kirkup, Piers J. Ingram, Konrad H. Paszkiewicz, Eric Ng, Zhihao Wu, Siyu Guan, Marvin M. Taguiam, Vicente Sancenon, and Dipti Thakkar
- Abstract
In recent years, HER3 has increasingly been implicated in the progression of a variety of tumor types and in acquired resistance to EGFR and HER2 therapies. Whereas EGFR and HER2 primarily signal through the MAPK pathway, HER3, as a heterodimer with EGFR or HER2, potently activates the PI3K pathway. Despite its critical role, previous attempts to target HER3 with neutralizing antibodies have shown disappointing efficacy in the clinic, most likely due to suboptimal and indirect mechanisms of action that fail to completely block heterodimerization; for example, tumors can escape inhibition of ligand binding by upregulating ligand-independent mechanisms of HER3 activation. We therefore developed 10D1F, a picomolar affinity, highly specific anti-HER3 neutralizing antibody that binds the HER3 heterodimerization interface, a region that was hitherto challenging to raise antibodies against. We demonstrate that 10D1F potently inhibits both EGFR:HER3 and HER2:HER3 heterodimerization to durably suppress activation of the PI3K pathway in a broad panel of tumor models. Even as a monotherapy, 10D1F shows superior inhibition of tumor growth in the same cell lines both in vitro and in mouse xenograft experiments, when compared with other classes of anti-HER3 antibodies. This includes models demonstrating ligand-independent activation of heterodimerization as well as constitutively activating mutations in the MAPK pathway. Possessing favorable pharmacokinetic and toxicologic profiles, 10D1F uniquely represents a new class of anti-HER3 neutralizing antibodies with a novel mechanism of action that offers significant potential for broad clinical benefit.10D1F is a novel anti-HER3 antibody that uniquely binds the receptor dimerization interface to block ligand-dependent and independent heterodimerization with EGFR/HER2 and thus more potently inhibits tumor growth than existing anti-HER3 antibodies.
- Published
- 2023
12. Supplementary Figure 2 from 10D1F, an Anti-HER3 Antibody That Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Jerome D. Boyd-Kirkup, Piers J. Ingram, Konrad H. Paszkiewicz, Eric Ng, Zhihao Wu, Siyu Guan, Marvin M. Taguiam, Vicente Sancenon, and Dipti Thakkar
- Abstract
10D1F interacts with linear peptides from the HER3 domain II dimerization interface
- Published
- 2023
13. Supplemental Table 3 from 10D1F, an Anti-HER3 Antibody That Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Jerome D. Boyd-Kirkup, Piers J. Ingram, Konrad H. Paszkiewicz, Eric Ng, Zhihao Wu, Siyu Guan, Marvin M. Taguiam, Vicente Sancenon, and Dipti Thakkar
- Abstract
Supplemental Table 3 - Toxicological data showing no adverse effects of 10D1F on the indicated hematological and biochemistry parameters in multiple rodent models
- Published
- 2023
14. Supplemental Table 1 from 10D1F, an Anti-HER3 Antibody That Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Jerome D. Boyd-Kirkup, Piers J. Ingram, Konrad H. Paszkiewicz, Eric Ng, Zhihao Wu, Siyu Guan, Marvin M. Taguiam, Vicente Sancenon, and Dipti Thakkar
- Abstract
Supplemental Table 1 - Summary of the published functional mechanisms of action for anti-HER3 neutralizing antibodies in development
- Published
- 2023
15. 532 Anti-VISTA antibody HMBD-002 reprograms tumour associated macrophages and promotes cytotoxic T cell response
- Author
-
Bhushan Dharmadhikari, Dipti Thakkar, Olga Zharkova, Debleena Ray, Jason Lai, Konrad Paszkiewicz, Piers Ingram, and Jerome Boyd-Kirkup
- Published
- 2022
16. The Fc-mediated effector functions of a potent SARS-CoV-2 neutralizing antibody, SC31, isolated from an early convalescent COVID-19 patient, are essential for the optimal therapeutic efficacy of the antibody
- Author
-
Chien-Te K Tseng, Dipti Thakkar, Boyd-Kirkup Jerome Douglas, Trevor Brasel, Paul A. MacAry, Dong Ling Wang, Jason E. Comer, Hannes Hentze, Ingram Piers, Brendon J. Hanson, Angeline P.C. Lim, Guan Siyu, Maricela Torres, Kiren Purushotorman, Damian O’Connell, Jacklyn J.Y. Neo, Guo Hui Teo, Steven K.K. Wong, Conrad E.Z. Chan, Shirley Gek Kheng Seah, Venkateshan S. Prativadibhayankara, Paul Edward Hutchinson, De Hoe Chye, Jenny G. Low, Gary S.L. Loh, Kantharaj Ethirajulu, David C. Lye, Jie Hui Lim, Pui San Wong, Barnaby Edward Young, Alan D.T. Barrett, and Shane Massey
- Subjects
Genetically modified mouse ,biology ,business.industry ,Epitope ,Immune system ,In vivo ,Immunology ,biology.protein ,Medicine ,Antibody-dependent enhancement ,Antibody ,business ,Neutralizing antibody ,Viral load - Abstract
SARS-CoV-2-neutralizing antibodies are promising therapeutics for COVID-19. However, little is known about the mechanisms of action of these antibodies or their effective dosing windows. We report the discovery and development of SC31, a potent SARS-CoV-2 neutralizing IgG1 antibody, originally isolated from a convalescent patient at day 27 after the onset of symptoms. Neutralization occurs via a binding epitope that maps within the ACE2 interface of the SARS-CoV-2 Spike protein, conserved across all common circulating SARS-CoV-2 mutants. In SARS-CoV-2 infected K18-human ACE2 transgenic mice, SC31 demonstrated potent survival benefit by dramatically reducing viral load concomitant with attenuated pro-inflammatory responses linked to severe systemic disease, such as IL-6. Comparison with a Fc-null LALA variant of SC31 demonstrated that optimal therapeutic efficacy of SC31 requires intact Fc-mediated effector functions that can further induce an IFNγ-driven anti-viral immune response. Dose-dependent efficacy for SC31 was observed down to 5mg/kg when dosed before the activation of lung inflammatory responses. Importantly, despite FcγR binding, no evidence of antibody dependent enhancement was observed with the Fc-competent SC31 even at sub-therapeutic doses. Therapeutic efficacy was confirmed in SARS-CoV-2-infected hamsters, where SC31 again significantly reduced viral load, decreased lung lesions and inhibited progression to severe disease manifestations. This study underlines the potential for significant COVID-19 patient benefit for the SC31 antibody that justifies rapid advancement to the clinic, as well as highlighting the importance of appropriate mechanistic and functional studies during development.One Sentence SummaryAnti-SARS-CoV-2 IgG1 antibody SC31 controls infectionin vivoby blocking SP:ACE2 binding and triggering a Fc-mediated anti-viral response.
- Published
- 2020
17. Abstract P197: An anti-HER3 antibody, HMBD-001, that uniquely binds to and blocks the HER3 heterodimerization interface, shows superior tumor growth inhibition in biomarker-defined preclinical cancer models including NRG1-fusion driven cancers
- Author
-
Dipti Thakkar, Shalini Paliwal, Shreya Kar, Namita Gandhi, Konrad Paszkiewicz, Piers Ingram, and Jerome Boyd-Kirkup
- Subjects
Cancer Research ,Oncology - Abstract
HER3 activation, through NRG1 ligand-dependent and independent heterodimerization with HER2 or EGFR, can drive tumor growth and survival via potent PI3K pathway signaling. HER3 activation has emerged as an important mechanism for both tumor progression and acquired resistance to standard of care therapies in multiple indications. With an effective biomarker strategy to select for patients with HER3 driven cancer, potent inhibition of HER3-driven heterodimerization has the potential to confer profound clinical impact. HER3 targeting approaches to date have not shown expected clinical efficacy. Suboptimal HER3 inhibition is one possible explanation; to prevent downstream PI3K signalling, it is critical to fully block both ligand-dependent and independent HER3 activation. Further, despite the clinical evaluation of several anti-HER3 antibodies, only limited progress has been made to establish predictive biomarkers of response to HER3 inhibition. The most thoroughly studied biomarkers predicting response have been increased expression and genomic rearrangements of the HER3 ligand, NRG1. For example, oncogenic NRG1-fusions have been identified as tumor drivers in up to 0.2% of all solid cancers. Few biomarkers predicting lack of response have been reported to date. HMBD-001, is an unique anti-HER3 antibody rationally developed to bind the dimerization interface of HER3 in order to block all HER3 heterodimerization. In contrast to other anti-HER3 antibodies that predominantly bind to the NRG1 binding domain, we observed potent and superior in vitro and in vivo tumor growth inhibition for HMBD-001 in multiple cancer models, including NRG1-fusion driven models. We show that HMBD-001 binds to HER3 with high affinity even in the presence of high concentrations of NRG1, in contrast to other antibodies in which NRG1 competes for the HER3 ligand binding site, thereby decreasing affinity and likely potency. Notably, HMBD-001 treatment of an NRG1-fusion ovarian PDX model showed superior tumor growth inhibition compared with anti-HER3 and anti-HER2/HER3 bispecific antibodies targeting the NRG1 binding domain of HER3. To develop a robust biomarker signature for patient selection beyond NRG1-fusions, we interrogated the genomic and transcriptomic data of all preclinical models for which HMBD-001 efficacy data was available, including in vitro and in vivo models representing multiple HER3 associated cancer sub-types. We identified a novel gene signature that was robustly predictive of HMBD-001 response across a wide range of cancers, including loss of function and gain of function mutations in specific downstream mediators of the MAPK and PI3K signaling pathways. In conclusion, the superior potency of HMBD-001 in preclinical models predicts more complete inhibition of HER3 and better responses in HER3 driven cancers that can be identified using a novel gene signature biomarker. Clinical trials of HMBD-001 in HER3 driven cancers, including those with NRG1-fusions, are expected to commence in 2021. Citation Format: Dipti Thakkar, Shalini Paliwal, Shreya Kar, Namita Gandhi, Konrad Paszkiewicz, Piers Ingram, Jerome Boyd-Kirkup. An anti-HER3 antibody, HMBD-001, that uniquely binds to and blocks the HER3 heterodimerization interface, shows superior tumor growth inhibition in biomarker-defined preclinical cancer models including NRG1-fusion driven cancers [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2021 Oct 7-10. Philadelphia (PA): AACR; Mol Cancer Ther 2021;20(12 Suppl):Abstract nr P197.
- Published
- 2021
18. 469 A phase 1 first in human study of HMBD-002, an IgG4 monoclonal antibody targeting VISTA, as a monotherapy and combined with pembrolizumab in patients with advanced solid malignancies
- Author
-
Boyd-Kirkup Jerome Douglas, Leah DiMascio, Ingram Piers, Guan Siyu, Dipti Thakkar, Benjamin L. Musher, Eric K. Rowinsky, Jordi Rodon, Joseph Kim, Monica M. Mita, Alain C. Mita, and Joshua J. Gruber
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,Tumor microenvironment ,biology ,business.industry ,T cell ,Immunology ,Cancer ,Pembrolizumab ,medicine.disease ,Immune checkpoint ,Cytokine release syndrome ,medicine.anatomical_structure ,Internal medicine ,medicine ,biology.protein ,Molecular Medicine ,Immunology and Allergy ,Antibody ,business ,Triple-negative breast cancer - Abstract
BackgroundV-domain Ig suppressor of T cell Activation (VISTA), an immune checkpoint regulator predominantly expressed on myeloid cells, represents a promising therapeutic target due to its role in suppressing pro-inflammatory, anti-tumor responses within the tumor microenvironment (TME). Based on VISTA’s broad expression across immune cell subtypes, HMBD-002 has been designed as a non-depleting, IgG4 monoclonal antibody with high affinity and specificity to VISTA across species (human, cynomolgus monkey, and rodent) that has the ability to block a predicted counter-structure binding site. In preclinical studies, HMBD-002 significantly inhibited tumor growth, both as a monotherapy and in combination with pembrolizumab, while decreasing infiltration of suppressive myeloid cells within the TME and increasing T cell activity. While rapid serum clearance and immune toxicities (e.g. cytokine release syndrome) have been reported for IgG1 antibodies, these were not observed preclinically with HMBD-002. In addition to VISTA expression on pro-inflammatory immune cells, examination of VISTA expression across cancer types has revealed that several malignancies, particularly human samples of triple negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC), express high levels of VISTA, thereby providing a rationale for exploring these indications in clinical studies.MethodsThis Phase 1, first in human study is being conducted in two parts and will evaluate multiple doses and schedules of intravenously (IV) administered HMBD-002, with or without pembrolizumab, in patients with advanced solid tumors. Part 1 (dose escalation) seeks to identify the maximum tolerated dose (MTD), or the maximum tested dose, of HMBD-002 as a monotherapy, and separately, in combination with pembrolizumab to define the recommend doses for subsequent disease directed studies (i.e., recommended phase 2 dose [RP2D]). Part 2 (dose expansion) will assess the anti-cancer activity of HMBD-002 as a monotherapy at the RP2D in previously treated patients with TNBC, and NSCLC, and in combination with pembrolizumab in patients with TNBC, NSCLC, and other VISTA-expressing malignancies. The size of the disease-directed cohorts will be determined based on an interim futility analysis conducted upon enrollment of 15 patients. Safety, efficacy, pharmacokinetic, and pharmacodynamic endpoints will be monitored and reported. Correlative studies will assess pre- and post-treatment markers of immune activity in the periphery and the tumor microenvironment.AcknowledgementsThis work was funded in part by the Cancer Prevention and Research Institute of Texas (CPRIT).Ethics ApprovalThe study was approved by each participating Institution’s Institutional Review Board.
- Published
- 2021
19. The Fc-mediated effector functions of a potent SARS-CoV-2 neutralizing antibody, SC31, isolated from an early convalescent COVID-19 patient, are essential for the optimal therapeutic efficacy of the antibody
- Author
-
Jie Hui Lim, Pui San Wong, Steven K.K. Wong, Chien Te K. Tseng, Guan Siyu, Paul A. MacAry, Shane Massey, Shirley Gek Kheng Seah, Alan D.T. Barrett, Dipti Thakkar, Jenny G. Low, Guo Hui Teo, Kantharaj Ethirajulu, Jason E. Comer, Brendon J. Hanson, Boyd-Kirkup Jerome Douglas, Ingram Piers, Venkateshan S. Prativadibhayankara, Barnaby Edward Young, Trevor Brasel, Angeline P.C. Lim, Hannes Hentze, Maricela Torres, Dongling Wang, Kiren Purushotorman, Jacklyn J.Y. Neo, Conrad E.Z. Chan, De Hoe Chye, Paul Edward Hutchinson, and Gary S.L. Loh
- Subjects
RNA viruses ,Male ,0301 basic medicine ,Viral Diseases ,Chemokine ,Physiology ,Coronaviruses ,Cancer Treatment ,Monkeys ,Biochemistry ,Epitope ,Neutralization ,Immunoglobulin G ,Medical Conditions ,0302 clinical medicine ,Immune Physiology ,Cricetinae ,Chlorocebus aethiops ,Medicine and Health Sciences ,Medicine ,030212 general & internal medicine ,Enzyme-Linked Immunoassays ,Neutralizing antibody ,Pathology and laboratory medicine ,Mammals ,Immune System Proteins ,Multidisciplinary ,biology ,Eukaryota ,Medical microbiology ,Viral Load ,Infectious Diseases ,Oncology ,Viruses ,Vertebrates ,Spike Glycoprotein, Coronavirus ,Hamsters ,Cytokines ,Female ,Angiotensin-Converting Enzyme 2 ,SARS CoV 2 ,Pathogens ,Chemokines ,Antibody ,Viral load ,Macaque ,Research Article ,Primates ,SARS coronavirus ,Science ,Immunology ,Mice, Transgenic ,Research and Analysis Methods ,Microbiology ,Rodents ,Antibodies ,03 medical and health sciences ,Immune system ,Antibody Therapy ,Virology ,Old World monkeys ,Animals ,Humans ,Immunoassays ,Vero Cells ,Dose-Response Relationship, Drug ,SARS-CoV-2 ,business.industry ,Organisms ,Viral pathogens ,Biology and Life Sciences ,Proteins ,COVID-19 ,Covid 19 ,Convalescence ,Antibodies, Neutralizing ,Macaca mulatta ,Microbial pathogens ,Immunoglobulin Fc Fragments ,Disease Models, Animal ,030104 developmental biology ,Amniotes ,Immunologic Techniques ,biology.protein ,Clinical Immunology ,Clinical Medicine ,business ,Zoology ,Viral Transmission and Infection - Abstract
Although SARS-CoV-2-neutralizing antibodies are promising therapeutics against COVID-19, little is known about their mechanism(s) of action or effective dosing windows. We report the generation and development of SC31, a potent SARS-CoV-2 neutralizing antibody, isolated from a convalescent patient. Antibody-mediated neutralization occurs via an epitope within the receptor-binding domain of the SARS-CoV-2 Spike protein. SC31 exhibited potent anti-SARS-CoV-2 activities in multiple animal models. In SARS-CoV-2 infected K18-human ACE2 transgenic mice, treatment with SC31 greatly reduced viral loads and attenuated pro-inflammatory responses linked to the severity of COVID-19. Importantly, a comparison of the efficacies of SC31 and its Fc-null LALA variant revealed that the optimal therapeutic efficacy of SC31 requires Fc-mediated effector functions that promote IFNγ-driven anti-viral immune responses, in addition to its neutralization ability. A dose-dependent efficacy of SC31 was observed down to 5mg/kg when administered before viral-induced lung inflammatory responses. In addition, antibody-dependent enhancement was not observed even when infected mice were treated with SC31 at sub-therapeutic doses. In SARS-CoV-2-infected hamsters, SC31 treatment significantly prevented weight loss, reduced viral loads, and attenuated the histopathology of the lungs. In rhesus macaques, the therapeutic potential of SC31 was evidenced through the reduction of viral loads in both upper and lower respiratory tracts to undetectable levels. Together, the results of our preclinical studies demonstrated the therapeutic efficacy of SC31 in three different models and its potential as a COVID-19 therapeutic candidate.
- Published
- 2021
20. 10D1F, an Anti-HER3 Antibody that Uniquely Blocks the Receptor Heterodimerization Interface, Potently Inhibits Tumor Growth Across a Broad Panel of Tumor Models
- Author
-
Dipti Thakkar, Sancenon Vicente, Guan Siyu, Ingram Piers, Eric Ng, Paszkiewicz Konrad, Boyd-Kirkup Jerome Douglas, Wu Zhihao, and Marvin M. Taguiam
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Receptor, ErbB-3 ,Mice, Nude ,Rats, Sprague-Dawley ,03 medical and health sciences ,Epitopes ,Mice ,0302 clinical medicine ,Neoplasms ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,Receptor ,Neutralizing antibody ,PI3K/AKT/mTOR pathway ,Mice, Inbred BALB C ,biology ,Chemistry ,Xenograft Model Antitumor Assays ,In vitro ,Rats ,body regions ,030104 developmental biology ,Oncology ,Mechanism of action ,Cell culture ,030220 oncology & carcinogenesis ,Immunoglobulin G ,Cancer research ,biology.protein ,Female ,Antibody ,medicine.symptom - Abstract
In recent years, HER3 has increasingly been implicated in the progression of a variety of tumor types and in acquired resistance to EGFR and HER2 therapies. Whereas EGFR and HER2 primarily signal through the MAPK pathway, HER3, as a heterodimer with EGFR or HER2, potently activates the PI3K pathway. Despite its critical role, previous attempts to target HER3 with neutralizing antibodies have shown disappointing efficacy in the clinic, most likely due to suboptimal and indirect mechanisms of action that fail to completely block heterodimerization; for example, tumors can escape inhibition of ligand binding by upregulating ligand-independent mechanisms of HER3 activation. We therefore developed 10D1F, a picomolar affinity, highly specific anti-HER3 neutralizing antibody that binds the HER3 heterodimerization interface, a region that was hitherto challenging to raise antibodies against. We demonstrate that 10D1F potently inhibits both EGFR:HER3 and HER2:HER3 heterodimerization to durably suppress activation of the PI3K pathway in a broad panel of tumor models. Even as a monotherapy, 10D1F shows superior inhibition of tumor growth in the same cell lines both in vitro and in mouse xenograft experiments, when compared with other classes of anti-HER3 antibodies. This includes models demonstrating ligand-independent activation of heterodimerization as well as constitutively activating mutations in the MAPK pathway. Possessing favorable pharmacokinetic and toxicologic profiles, 10D1F uniquely represents a new class of anti-HER3 neutralizing antibodies with a novel mechanism of action that offers significant potential for broad clinical benefit. 10D1F is a novel anti-HER3 antibody that uniquely binds the receptor dimerization interface to block ligand-dependent and independent heterodimerization with EGFR/HER2 and thus more potently inhibits tumor growth than existing anti-HER3 antibodies.
- Published
- 2019
21. HMBD-002 is a novel, neutralizing, anti-VISTA antibody exhibiting strong preclinical efficacy and safety, being developed as a monotherapy and in combination with pembrolizumab
- Author
-
Ingram Piers, Dipti Thakkar, Namita S. Gandhi, Leah DiMascio, Boyd-Kirkup Jerome Douglas, Guan Siyu, and Eric K. Rowinsky
- Subjects
Cancer Research ,Immune system ,Oncology ,biology ,business.industry ,biology.protein ,Cancer research ,Medicine ,Pembrolizumab ,Antibody ,business - Abstract
e14569 Background: Inhibition of immune checkpoints has been an effective strategy against several cancers, but efficacy gaps remain. VISTA is an immunoregulatory protein expressed on both hematopoietic and tumor cells and has been shown to induce an immunosuppressive environment by repressing T-cell activation and cytokine production. VISTA expression has been shown to be upregulated following treatment with immune checkpoint inhibitors. Hummingbird Bioscience is developing HMBD-002, an IgG4 antibody against VISTA with high binding specificity across species (human, cyno and rodent) and low pM affinity to the region of VISTA required for engagement with interaction partners, including VSIG3. As HMBD-002 is an IgG4 isotype, it uniquely blocks the inhibitory function of VISTA and releases immune suppression without depleting VISTA+ cells. Here we establish the preclinical efficacy and safety of HMBD-002 along with the rationale for development of HMBD-002 as a monotherapy and in combination with PD1 inhibitors in triple negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC). Methods: The effect of HMBD-002 in an in vitro model of human immune activation was evaluated using an allogenic Mixed Lymphocyte Reaction (MLR) assay. Tumor growth inhibition studies were conducted in syngeneic murine cell-derived xenograft (CDX) models. The PK and tolerability of HMBD-002 was evaluated in rats and cynomolgus monkeys with a 21 or 28-day observation period. The expression profile of VISTA was examined in different cancers by IHC staining of FFPE TMAs of various solid cancers. Results: HMBD-002 induced a significant dose dependent increase in the levels of inflammatory cytokines, IFN-γ and TNF- α and suppressed the expansion of CD14+ monocytes. Treatment with HMBD-002 significantly inhibited tumor growth and improved survival in CDX models. HMBD-002 is well tolerated in rats and cynomolgus monkeys with acceptable serum half-life, no HMBD-002 related morbidity/mortality or clinical signs, and no HMBD-002 related changes were observed in body weight, food consumption, clinical chemistry, or hematology parameters. HMBD-002 did not induce cytokine release in human PBMCs or whole blood. TNBC and NSCLC patients were found to harbor the highest VISTA expression across cancer types tested, providing a rationale for exploring these indications in upcoming clinical studies. Conclusions: These studies provide the rationale for development of HMBD-002 as a monotherapy and in combination with pembrolizumab to treat patients with solid tumors expressing VISTA in the tumor microenvironment. A Phase 1 study will be initiated in 2021.
- Published
- 2021
22. Considerations for treatment duration in responders to immune checkpoint inhibitors
- Author
-
Jiajia Zhang, Dipti Thakkar, Vaia Florou, Rachel Howard, Rania H. Younis, Esha Sachdev, Aideen E. Ryan, Christopher A. Fuhrman, Rosa Nguyen, Abigail E. Overacre-Delgoffe, Sabina Kaczanowska, Maria E. Rodriguez-Ruiz, Thomas U. Marron, Daniel J Olson, Jennifer L. Guerriero, Todd Bartkowiak, Jessica E. Thaxton, Sarah E. Church, David H. Aggen, Michal Sheffer, Ravi Patel, Sangeetha M. Reddy, Kristin G. Anderson, and Abdul Rafeh Naqash
- Subjects
Cancer Research ,medicine.medical_specialty ,Time Factors ,medicine.medical_treatment ,Treatment duration ,Immune checkpoint inhibitors ,Immunology ,Review ,Risk Assessment ,Drug Administration Schedule ,03 medical and health sciences ,0302 clinical medicine ,Costimulatory and Inhibitory T-Cell Receptors ,Risk Factors ,Neoplasms ,Overall survival ,Humans ,Immunology and Allergy ,Medicine ,030212 general & internal medicine ,Intensive care medicine ,Immune Checkpoint Inhibitors ,Melanoma ,RC254-282 ,Pharmacology ,Clinical Trials as Topic ,Evidence-Based Medicine ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,Limiting ,Immunotherapy ,medicine.disease ,Clinical trial ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Patient Safety ,business - Abstract
Immune checkpoint inhibitors (ICIs) have improved overall survival for cancer patients, however, optimal duration of ICI therapy has yet to be defined. Given ICIs were first used to treat patients with metastatic melanoma, a condition that at the time was incurable, little attention was initially paid to how much therapy would be needed for a durable response. As the early immunotherapy trials have matured past 10 years, a significant per cent of patients have demonstrated durable responses; it is now time to determine whether patients have been overtreated, and if durable remissions can still be achieved with less therapy, limiting the physical and financial toxicity associated with years of treatment. Well-designed trials are needed to identify optimal duration of therapy, and to define biomarkers to predict who would benefit from shorter courses of immunotherapy. Here, we outline key questions related to health, financial and societal toxicities of over treating with ICI and present four unique clinical trials aimed at exposing criteria for early cessation of ICI. Taken together, there is a serious liability to overtreating patients with ICI and future work is warranted to determine when it is safe to stop ICI.
- Published
- 2021
23. A Genome-wide Association Study of Dupuytren Disease Reveals 17 Additional Variants Implicated in Fibrosis
- Author
-
Michael, Ng, Dipti, Thakkar, Lorraine, Southam, Paul, Werker, Roel, Ophoff, Kerstin, Becker, Michael, Nothnagel, Andre, Franke, Peter, Nürnberg, Ana Isabel, Espirito-Santo, David, Izadi, Hans Christian, Hennies, Jagdeep, Nanchahal, Eleftheria, Zeggini, and Dominic, Furniss
- Subjects
Dupuytren disease ,Genotype ,Gene Expression Profiling ,fibrosis ,Polymorphism, Single Nucleotide ,Article ,Cohort Studies ,Dupuytren Contracture ,Wnt Proteins ,hand surgery ,Case-Control Studies ,Proto-Oncogene Proteins ,Humans ,GWAS ,genetics ,Myofibroblasts ,Biomarkers ,Cells, Cultured ,Genome-Wide Association Study - Abstract
Individuals with Dupuytren disease (DD) are commonly seen by physicians and surgeons across multiple specialties. It is an increasingly common and disabling fibroproliferative disorder of the palmar fascia, which leads to flexion contractures of the digits, and is associated with other tissue-specific fibroses. DD affects between 5% and 25% of people of European descent and is the most common inherited disease of connective tissue. We undertook the largest GWAS to date in individuals with a surgically validated diagnosis of DD from the UK, with replication in British, Dutch, and German individuals. We validated association at all nine previously described signals and discovered 17 additional variants with p ≤ 5 × 10−8. As a proof of principle, we demonstrated correlation of the high-risk genotype at the statistically most strongly associated variant with decreased secretion of the soluble WNT-antagonist SFRP4, in surgical specimen-derived DD myofibroblasts. These results highlight important pathways involved in the pathogenesis of fibrosis, including WNT signaling, extracellular matrix modulation, and inflammation. In addition, many associated loci contain genes that were hitherto unrecognized as playing a role in fibrosis, opening up new avenues of research that may lead to novel treatments for DD and fibrosis more generally. DD represents an ideal human model disease for fibrosis research.
- Published
- 2017
24. Abstract 1729: Integrative immune profiling of syngeneic tumor models provides predictive immune signatures for treatment response with HMBD002, a novel anti-VISTA neutralizing antibody
- Author
-
Ingram Piers, Dipti Thakkar, Paszkiewicz Konrad, and Boyd-Kirkup Jerome Douglas
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,biology ,medicine.drug_class ,medicine.medical_treatment ,Immunotherapy ,Monoclonal antibody ,Epitope ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,Immune system ,Oncology ,medicine ,Cancer research ,biology.protein ,Antibody ,Neutralizing antibody - Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of immature myeloid cells with potent immune suppressive activity. Accumulation of pathologically activated MDSCs within the tumor microenvironment has emerged as an important mechanism of primary and acquired resistance to immunotherapy. MDSCs have high surface expression levels of V-domain immunoglobulin (Ig)-containing suppressor of T-cell activation (VISTA), a co-inhibitory negative checkpoint regulator (NCR). Immune suppression by VISTA+ MDSCs has been implicated in the failure of, and resistance to, anti-PD1/PDL1 therapy and represents a unique axis for NCR targeting in the non-responder population. Elucidating the dynamics of VISTA+ MDSCs within tumors is imperative for predicting response and patient stratification. HMBD-002 is a novel anti-VISTA neutralizing monoclonal antibody, developed using Hummingbird Bioscience's proprietary Rational Antibody Discovery platform, against a specific functional epitope on VISTA predicted to be involved in ligand binding. HMBD002 binds human, cyno and murine VISTA protein with high affinity (kD To investigate predictive immune signatures for HMBD002 antibody efficacy, as a monotherapy and in combination with anti- PD-L1 therapy, we established multiple mouse models of tumor immune subtypes using syngeneic cell lines. Tumor biopsies were analyzed before and during treatment, to determine the evolution of immune response and its effect on tumor burden within responders and non-responders. The combination of HMBD-002 and anti-PD-L1 was more effective than either antibody alone, especially in tumors that showed abundant MDSC infiltration. Immune phenotyping revealed that this was associated with a significant increase in activated CD8+ T effector cells and concomitant increase in IFN-gamma. Profiling syngeneic models for deeper insights into the interplay between the tumor, immune system and therapy, will facilitate the clinical development of immune checkpoint and combination therapies. Hummingbird Bioscience anticipates commencing First-in-Human trials of HMBD002 in 2019. Citation Format: Jerome D. Boyd-Kirkup, Dipti Thakkar, Konrad Paszkiewicz, Piers J. Ingram. Integrative immune profiling of syngeneic tumor models provides predictive immune signatures for treatment response with HMBD002, a novel anti-VISTA neutralizing antibody [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1729.
- Published
- 2018
25. Gene expression profiles of changes underlying different-sized human rotator cuff tendon tears
- Author
-
Zhidao Xia, Dipti Thakkar, Andrew Carr, Osnat Hakimi, and Salma Chaudhury
- Subjects
Matrix Metalloproteinase 3 ,Pathology ,medicine.medical_specialty ,Down-Regulation ,Matrix metalloproteinase ,Real-Time Polymerase Chain Reaction ,Rotator Cuff Injuries ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Medicine ,Humans ,Orthopedics and Sports Medicine ,Rotator cuff ,Aggrecan ,Retrospective Studies ,030222 orthopedics ,Metalloproteinase ,Principal Component Analysis ,business.industry ,Gene Expression Profiling ,Interleukins ,030229 sport sciences ,General Medicine ,musculoskeletal system ,Microarray Analysis ,Immunohistochemistry ,Matrix Metalloproteinases ,Tendon ,Up-Regulation ,ADAM Proteins ,medicine.anatomical_structure ,Case-Control Studies ,Tears ,Surgery ,business - Abstract
Background: Progressive cellular and extracellular matrix (ECM) changes related to age and disease severity have been demonstrated in rotator cuff tendon tears. Larger rotator cuff tears demonstrate structural abnormalities that potentially adversely influence healing potential. This study aimed to gain greater insight into the relationship of pathologic changes to tear size by analyzing gene expression profiles from normal rotator cuff tendons, small rotator cuff tears, and large rotator cuff tears. Methods: We analyzed gene expression profiles of 28 human rotator cuff tendons using microarrays representing the entire genome; 11 large and 5 small torn rotator cuff tendon specimens were obtained intraoperatively from tear edges, which we compared with 12 age-matched normal controls. We performed real-time polymerase chain reaction and immunohistochemistry for validation. Results: Torn rotator cuff tendons demonstrated upregulation of a number of key genes, such as matrix metalloproteinase 3, 10, 12, 13, 15, 21, and 25; a disintegrin and metalloproteinase (ADAM) 12, 15, and 22; and aggrecan. Amyloid was downregulated in all tears. Small tears displayed upregulation of bone morphogenetic protein 5. Chemokines and cytokines that may play a role in chemotaxis were altered; interleukins 3, 10, 13, and 15 were upregulated in tears, whereas interleukins 1, 8, 11, 18, and 27 were downregulated. Conclusions: The gene expression profiles of normal controls and small and large rotator cuff tear groups differ significantly. Extracellular matrix remodeling genes were found to contribute to rotator cuff tear pathogenesis. Rotator cuff tears displayed upregulation of a number of matrix metalloproteinase (3, 10, 12, 13, 15, 21, and 25), a disintegrin and metalloproteinase (ADAM 12, 15, and 22) genes, and downregulation of some interleukins (1, 8, and 27), which play important roles in chemotaxis. These gene products may potentially have a role as biomarkers of failure of healing or therapeutic targets to improve tendon healing.
- Published
- 2015
26. Abstract 587: HMBD002, a novel neutralizing antibody targeting a specific epitope on the co-inhibitory immune checkpoint receptor VISTA, displays potent anti-tumor effects in pre-clinical models
- Author
-
Ingram Piers, Dipti Thakkar, and Boyd-Kirkup Jerome Douglas
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,biology ,medicine.drug_class ,medicine.medical_treatment ,Immunotherapy ,Monoclonal antibody ,Molecular biology ,Epitope ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,Oncology ,medicine ,biology.protein ,Cancer research ,Antibody ,Neutralizing antibody ,Antigen-presenting cell - Abstract
V-domain immunoglobulin (Ig)-containing suppressor of T-cell activation (VISTA) also known as PD1 homolog (PD1H) is a co-inhibitory immune checkpoint receptor. VISTA is predominantly expressed on hematopoietic cells, particularly myeloid derived suppressor cells and antigen presenting cells, and at lower levels on CD4+ and CD8+ T cells and Foxp3 Tregs. Multiple studies have demonstrated VISTA can strongly suppress human T-cell activation, and the presence of high VISTA expressing cells in the tumor microenvironment has been postulated as playing a critical role in tumorigenesis and resistance to immunotherapy. VISTA neutralizing antibodies have been previously observed in pre-clinical models to increase the abundance of tumor infiltrating effector T cells as well as their effector functions, resulting in enhanced control of tumor growth, even in the absence of detectable expression of VISTA on the tumor cells, a potential advantage over PD1 or PDL1 blockade. There is an urgent need to develop the most clinically effective anti-VISTA antibodies however the limited characterization of the VISTA pathway and the uncertainty around the cognate VISTA ligand has limited the extent to which in vitro studies can support the selection of optimal antibodies for development. Hummingbird Bioscience’s proprietary Rational Antibody Development platform to the design of neutralizing antibodies against specific epitopes on VISTA. Extensive in silico analyses of the VISTA structure and comparative structural modeling against other B7 protein family members has enabled the prediction of key binding sites where antibody binding will inhibit function, while preserving necessary safety and developability profiles. Subsequently, a directed evolution strategy was used to isolate monoclonal antibodies specifically targeting these key predicted functional domains on the native folded VISTA protein. Hummingbird’s anti-VISTA antibodies bind human and murine VISTA protein with high affinity (kD 60% inhibition of tumor growth (after 15 days) with no observable toxicity. Further in vitro and in vivo validation experiments in other cancer models are ongoing, both as monotherapy and in combination with other immune therapies. The first in-human trial of HMBD-002 is planned for 2018. Citation Format: Piers J. Ingram, Dipti Thakkar, Jerome D. Boyd-Kirkup. HMBD002, a novel neutralizing antibody targeting a specific epitope on the co-inhibitory immune checkpoint receptor VISTA, displays potent anti-tumor effects in pre-clinical 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 587. doi:10.1158/1538-7445.AM2017-587
- Published
- 2017
27. Abstract 24: HMBD001, a novel anti-ErbB3 antibody with a unique mechanism of action, effectively inhibits tumor growth in pre-clinical models of ErbB3+ solid tumors
- Author
-
Dipti Thakkar, Ingram Piers, and Boyd-Kirkup Jerome Douglas
- Subjects
Cancer Research ,biology ,medicine.drug_class ,Chemistry ,Cancer ,Pharmacology ,Monoclonal antibody ,Ligand (biochemistry) ,medicine.disease ,Epitope ,Oncology ,Mechanism of action ,biology.protein ,medicine ,ERBB3 ,Antibody ,medicine.symptom ,PI3K/AKT/mTOR pathway - Abstract
Gastric cancer is the second highest cause of cancer mortality in Asia. Over-expression of ErbB3, a cell surface RTK, occurs in ~60% of mGC patients where it is significantly associated with poorer survival. Ligand binding to ErbB3 triggers heterodimerization and downstream signaling through Akt. Neutralizing antibodies that inhibit ligand binding show poor efficacy in clinical trials, likely due to high levels of ligand independent activation in many tumor types. Directly inhibiting heterodimerization could be more effective, however, current methods of antibody isolation offer limited control over the site of antibody binding and, therefore, mechanism of action, requiring extensive screening of antibodies and posing significant challenges to identifying suitable candidates for therapeutic development. Hummingbird Bioscience has used its proprietary Rational Antibody Development Platform for the design and development of novel therapeutic antibodies against ErbB3. Computational sequence and structural analyses predicted highly specific surface epitopes, with strong antigenicity and good safety profiles, where antibody binding would inhibit dimerization. Subsequently, Hummingbird’s directed evolution mouse immunization strategy efficiently isolated monoclonal antibodies that bound with low nM (Kd < 5nM) affinity to these epitopes on native folded ErbB3. Furthermore, these mAbs showed ELISA cross-reactivity to ErbB3 in model organisms (mouse, rat, monkey) but no binding to other EGFR family proteins. Antibody HMBD001 was found to strongly inhibit the phosphorylation of ErbB3 and decrease downstream signaling through the Akt pathway (90% decrease in p-ErbB3 and 60% decrease in p-AKT observed at 24 hrs) resulting in the inhibition of tumor cell proliferation in models of ErbB3+ gastric cancer (over 90% inhibition of NCI-N87 proliferation after 5 days) and breast cancer (up to 85% inhibition of BT474 proliferation after 5 days). In mouse NCI-N87 gastric cancer CDX models, weekly doses of HMBD001 achieved greater than 90% inhibition of tumor growth (at 25 days) with no observable adverse toxicity. The first in-human trial of HMBD001 is planned for 2018. Citation Format: Jerome D. Boyd-Kirkup, Dipti Thakkar, Piers J. Ingram. HMBD001, a novel anti-ErbB3 antibody with a unique mechanism of action, effectively inhibits tumor growth in pre-clinical models of ErbB3+ solid tumors [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 24. doi:10.1158/1538-7445.AM2017-24
- Published
- 2017
28. Distribution and expression of type VI collagen and elastic fibers in human rotator cuff tendon tears
- Author
-
Dipti Thakkar, Andrew Carr, Tyler Grant, and Osnat Hakimi
- Subjects
Pathology ,medicine.medical_specialty ,Fibrillin-1 ,macromolecular substances ,Degeneration (medical) ,Collagen Type VI ,Matrix (biology) ,Fibrillins ,Real-Time Polymerase Chain Reaction ,Biochemistry ,Rotator Cuff Injuries ,Extracellular matrix ,Rotator Cuff ,Rheumatology ,medicine ,Humans ,Orthopedics and Sports Medicine ,Rotator cuff ,Molecular Biology ,DNA Primers ,Analysis of Variance ,Extracellular Matrix Proteins ,Microscopy, Confocal ,biology ,Chemistry ,Gene Expression Profiling ,Microfilament Proteins ,Cell Biology ,Anatomy ,musculoskeletal system ,Elastic Tissue ,Immunohistochemistry ,Tendon ,medicine.anatomical_structure ,biology.protein ,Tears ,Elastin ,Fibrillin - Abstract
There is increasing evidence for a progressive extracellular matrix change in rotator cuff disease progression. Directly surrounding the cell is the pericellular matrix, where assembly of matrix aggregates typically occurs making it critical in the response of tendon cells to pathological conditions. Studies in animal models have identified type VI collagen, fibrillin-1 and elastin to be located in the pericellular matrix of tendon and contribute in maintaining the structural and biomechanical integrity of tendon. However, there have been no reports on the localization of these proteins in human tendon biopsies. This study aimed to characterize the distribution of these ECM components in human rotator cuffs and gain greater insight into the relationship of pathology to tear size by analyzing the distribution and expression profiles of these ECM components. Confocal microscopy confirmed the localization of these structural molecules in the pericellular matrix of the human rotator cuff. Tendon degeneration led to an increased visibility of these components with a significant disorganization in the distribution of type VI collagen. At the genetic level, an increase in tear size was linked to an increased transcription of type VI collagen and fibrillin-1 with no significant alteration in the elastin levels. This is the first study to confirm the localization of type VI collagen, elastin and fibrillin-1 in the pericellular region of human supraspinatus tendon and assesses the effect of tendon degeneration on these structures, thus providing a useful insight into the composition of human rotator cuff tears which can be instrumental in predicting disease prognosis.
- Published
- 2014
29. Proteomic studies coupled with RNAi methodologies can shed further light on the downstream effects of telomerase in glioma
- Author
-
Leroy Alexander Shervington, Amal Shervington, and Dipti Thakkar
- Subjects
Proteomics ,Cancer Research ,Telomerase ,Vimentin ,Biology ,Downregulation and upregulation ,RNA interference ,Glioma ,Cell Line, Tumor ,Gene expression ,medicine ,Humans ,Telomerase reverse transcriptase ,Brain Neoplasms ,General Medicine ,medicine.disease ,Neoplasm Proteins ,Oncology ,Tumor progression ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Cancer research ,biology.protein ,RNA Interference - Abstract
A comprehensive proteomic study utilizing 2D-DIGE and MALDI-TOF was used to assess the effect of inhibiting two different regulatory mechanisms of telomerase in glioma. RNAi was used to target hTERT and hsp90α. Inhibition of telomerase activity resulted in downregulation of various cytoskeletal proteins with correlative evidence of the involvement of telomerase in regulating the expression of vimentin. Inhibition of telomerase via sihTERT resulted in the downregulation of vimentin expression in glioma cell lines in a grade-specific manner. This study identified novel downstream role of telomerase in regulating the expression of vimentin, thereby affecting tumor progression and metastasis.
- Published
- 2011
30. Can hsp90alpha-targeted siRNA combined with TMZ be a future therapy for glioma?
- Author
-
Rahima Patel, Nichola Cruickshanks, Amal Shervington, Leroy Alexander Shervington, Dipti Thakkar, and Chinmay Munje
- Subjects
Cancer Research ,Time Factors ,Cell Survival ,medicine.medical_treatment ,Pharmacology ,Transfection ,Inhibitory Concentration 50 ,RNA interference ,Glioma ,Cell Line, Tumor ,Temozolomide ,Gene silencing ,Medicine ,Humans ,HSP90 Heat-Shock Proteins ,RNA, Messenger ,RNA, Small Interfering ,Antineoplastic Agents, Alkylating ,Chemotherapy ,Dose-Response Relationship, Drug ,business.industry ,Brain Neoplasms ,General Medicine ,Genetic Therapy ,medicine.disease ,Dacarbazine ,Gene Expression Regulation, Neoplastic ,Oncology ,Apoptosis ,Cell culture ,Chemotherapy, Adjuvant ,Cancer research ,RNA Interference ,Cisplatin ,business ,medicine.drug - Abstract
Hsp90alpha's vital role in cell cycle progression and apoptosis together with its presence in gliomas and absence in normal tissue, make it a credible target for cancer therapy. Three sets of dsRNA oligos designed to align different regions of the hsp90alpha sequence were used to downregulate hsp90alpha. SiRNA 1, 2, and 3 resulted in significant levels of silencing of hsp90alpha after 48 hr treatment (p.0001). Concurrent treatment of the glioma cell line U87-MG with siRNA 1 and temozolomide (TMZ) resulted in a 13-fold reduction in the dose of TMZ required to achieve a similar effect if TMZ was used alone.
- Published
- 2010
31. Proteomics: the tool to bridge the gap between the facts and fables of telomerase
- Author
-
Amal Shervington and Dipti Thakkar
- Subjects
Proteomics ,Cancer Research ,Telomerase ,Diagnostic marker ,Genomics ,Computational biology ,Biology ,Bioinformatics ,Prognosis ,Gene Expression Regulation, Enzymologic ,Epigenesis, Genetic ,Gene Expression Regulation, Neoplastic ,Neoplasms ,Biomarkers, Tumor ,Animals ,Humans ,Protein Binding - Abstract
Genomic analysis has provided a huge cohort of data that has shed light on the regulation and structure of telomerase and will continue to influence its study profoundly. However, genomic and proteomic data need to be collated in order to bridge the gap between telomerase regulation and its crucial role in carcinogenesis. This review highlights significant advances in understanding the regulation of telomerase activity in the realm of genomics and proteomics focusing on the importance of telomerase as a future prognostic and diagnostic marker.
- Published
- 2009
32. The sensitization of glioma cells to cisplatin and tamoxifen by the use of catechin
- Author
-
Rahima Patel, Dipti Thakkar, Sharad Menon, Vidya Pawar, and Amal Shervington
- Subjects
Telomerase ,Cell Survival ,Antineoplastic Agents ,Pharmacology ,Epigallocatechin gallate ,complex mixtures ,Catechin ,chemistry.chemical_compound ,Glioma ,Cell Line, Tumor ,Genetics ,medicine ,Cytotoxic T cell ,Humans ,Molecular Biology ,Sensitization ,Cisplatin ,Cell Death ,food and beverages ,General Medicine ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Tamoxifen ,medicine.anatomical_structure ,chemistry ,Drug Screening Assays, Antitumor ,medicine.drug - Abstract
Telomerase expression strongly correlates with the grade of malignancy in glioma with inhibition illustrating a definite increase in chemosensitivity. This study was designed to investigate the effects of a green tea derivative, epigallocatechin-3-gallate (EGCG); together with either cisplatin or tamoxifen in glioma, and to investigate whether these effects are mediated through telomerase suppression. EGCG showed a significant cytotoxic effect on 1321N1 cells after 24 h and on U87-MG cells after 72 h (P < 0.001) without significantly affecting the normal astrocytes. Treatment with EGCG inhibited telomerase expression significantly (P < 0.01) and enhanced the effect of cisplatin and tamoxifen in both 1321N1 (P < 0.01) and U87-MG (P < 0.001) cells. EGCG, as a natural product has enormous potential to be an anti-cancer agent capable of enhancing tumour cell sensitivity to therapy.
- Published
- 2008
33. Ascorbic acid is essential for significant collagen deposition by human tenocytes in vitro
- Author
-
Osnat Hakimi, Andrew Carr, Clarence Yapp, Dipti Thakkar, and Raewyn Poulson
- Subjects
Vitamin C ,Tissue engineering ,Biochemistry ,Tendon cell ,Cell growth ,Chemistry ,Context (language use) ,Ascorbic acid ,Cell morphology ,In vitro - Abstract
Objective: We sought to compare L-ascorbic acid (AA), the natural form of vitamin C, and magnesium ascorbyl phosphate (MAP), a stable derivative, for their effect on cell proliferation, culture collagen content and collagen expression in tendon-derived cells. Methods: Tenocytes received these two ascorbate formulations at concentrations between 0.01 mM and 2 M. The effect of supplementation was evaluated in terms of cell growth, collagen expression, collagen secretion, cell morphology and collagen localization. Results: MAP supplementation resulted in dose-dependent increase in growth rates, whilst AA supplementation was toxic to tendon-derived cells from as low as 1 mM. Collagen expression was only mildly modulated by ascorbic acid, but collagen accumulation was significantly increased, supporting the hypothesis of its possible role in secretion. Immunofluorescence staining revealed differential localization of collagen I in vitro with collagen localized outside cells in the presence of ascorbic acid. Multi-photon microscopy further demonstrated that collagen fibrils were only formed in the presence of ascorbic acid. Conclusions: Taken together, these results suggest that ascorbic acid, preferably the magnesium ascorbyl phosphate formulation, should be routinely used as a supplement to tendon cell culture when collagen fibrils are required, especially in the context of tissue engineering and scaffold development.
- Published
- 2014
34. The sensitization of glioma cells to cisplatin and tamoxifen by the use of catechin.
- Author
-
Amal Shervington, Vidya Pawar, Sharad Menon, Dipti Thakkar, and Rahima Patel
- Abstract
Abstract Telomerase expression strongly correlates with the grade of malignancy in glioma with inhibition illustrating a definite increase in chemosensitivity. This study was designed to investigate the effects of a green tea derivative, epigallocatechin-3-gallate (EGCG); together with either cisplatin or tamoxifen in glioma, and to investigate whether these effects are mediated through telomerase suppression. EGCG showed a significant cytotoxic effect on 1321N1 cells after 24 h and on U87-MG cells after 72 h (P P P P [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.