Network


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

Hotspot


Dive into the research topics where Sher Karki is active.

Publication


Featured researches published by Sher Karki.


Blood | 2012

Potent in vitro and in vivo activity of an Fc-engineered humanized anti-HM1.24 antibody against multiple myeloma via augmented effector function

Yu-Tzu Tai; Holly M. Horton; Sun-Young Kong; Erik Pong; Hsing Chen; Saso Cemerski; Matthew J. Bernett; Duc-Hanh T. Nguyen; Sher Karki; Seung Y. Chu; Greg A. Lazar; Nikhil C. Munshi; John R. Desjarlais; Kenneth C. Anderson; Umesh Muchhal

HM1.24, an immunologic target for multiple myeloma (MM) cells, has not been effectively targeted with therapeutic monoclonal antibodies (mAbs). In this study, we investigated in vitro and in vivo anti-MM activities of XmAb5592, a humanized anti-HM1.24 mAb with Fc-domain engineered to significantly enhance FcγR binding and associated immune effector functions. XmAb5592 increased antibody-dependent cellular cytotoxicity (ADCC) several fold relative to the anti-HM1.24 IgG1 analog against both MM cell lines and primary patient myeloma cells. XmAb5592 also augmented antibody dependent cellular phagocytosis (ADCP) by macrophages. Natural killer (NK) cells became more activated by XmAb5592 than the IgG1 analog, evidenced by increased cell surface expression of granzyme B-dependent CD107a and MM cell lysis, even in the presence of bone marrow stromal cells. XmAb5592 potently inhibited tumor growth in mice bearing human MM xenografts via FcγR-dependent mechanisms, and was significantly more effective than the IgG1 analog. Lenalidomide synergistically enhanced in vitro ADCC against MM cells and in vivo tumor inhibition induced by XmAb5592. A single dose of 20 mg/kg XmAb5592 effectively depleted both blood and bone marrow plasma cells in cynomolgus monkeys. These results support clinical development of XmAb5592, both as a monotherapy and in combination with lenalidomide, to improve patient outcome of MM.


Molecular Cancer Therapeutics | 2015

AGS67E, an Anti-CD37 Monomethyl Auristatin E Antibody–Drug Conjugate as a Potential Therapeutic for B/T-Cell Malignancies and AML: A New Role for CD37 in AML

Daniel S. Pereira; Claudia I. Guevara; Liqing Jin; Nathan Mbong; Alla Verlinsky; Ssucheng J. Hsu; Hector Aviña; Sher Karki; Joseph D. Abad; Sung-Ju Moon; Faisal Malik; Michael Y. Choi; Zili An; Kendall Morrison; Pia M. Challita-Eid; Fernando Doñate; Ingrid B.J.K. Joseph; Thomas J. Kipps; John E. Dick; David R. Stover

CD37 is a tetraspanin expressed on malignant B cells. Recently, CD37 has gained interest as a therapeutic target. We developed AGS67E, an antibody–drug conjugate that targets CD37 for the potential treatment of B/T-cell malignancies. It is a fully human monoclonal IgG2 antibody (AGS67C) conjugated, via a protease-cleavable linker, to the microtubule-disrupting agent monomethyl auristatin E (MMAE). AGS67E induces potent cytotoxicity, apoptosis, and cell-cycle alterations in many non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia (CLL) cell lines and patient-derived samples in vitro. It also shows potent antitumor activity in NHL and CLL xenografts, including Rituxan-refractory models. During profiling studies to confirm the reported expression of CD37 in normal tissues and B-cell malignancies, we made the novel discovery that the CD37 protein was expressed in T-cell lymphomas and in AML. AGS67E bound to >80% of NHL and T-cell lymphomas, 100% of CLL and 100% of AML patient-derived samples, including CD34+CD38− leukemic stem cells. It also induced cytotoxicity, apoptosis, and cell-cycle alterations in AML cell lines and antitumor efficacy in orthotopic AML xenografts. Taken together, this study shows not only that AGS67E may serve as a potential therapeutic for B/T-cell malignancies, but it also demonstrates, for the first time, that CD37 is well expressed and a potential drug target in AML. Mol Cancer Ther; 14(7); 1650–60. ©2015 AACR.


Cancer Research | 2016

Enfortumab Vedotin Antibody-Drug Conjugate Targeting Nectin-4 is a Highly Potent Therapeutic Agent in Multiple Preclinical Cancer Models

Pia M. Challita-Eid; Daulet Satpayev; Zili An; Kendall Morrison; Yuriy Shostak; Arthur B. Raitano; R. Nadell; Wendy Liu; Dawn Ratay Lortie; Linnette Capo; Alla Verlinsky; Monica Leavitt; Faisal Malik; Hector Aviña; Claudia I. Guevara; N. Dinh; Sher Karki; Banmeet Anand; Daniel S. Pereira; Ingrid B.J.K. Joseph; Fernando Doñate; David R. Stover

The identification of optimal target antigens on tumor cells is central to the advancement of new antibody-based cancer therapies. We performed suppression subtractive hybridization and identified nectin-4 (PVRL4), a type I transmembrane protein and member of a family of related immunoglobulin-like adhesion molecules, as a potential target in epithelial cancers. We conducted immunohistochemical analysis of 2,394 patient specimens from bladder, breast, lung, pancreatic, ovarian, head/neck, and esophageal tumors and found that 69% of all specimens stained positive for nectin-4. Moderate to strong staining was especially observed in 60% of bladder and 53% of breast tumor specimens, whereas the expression of nectin-4 in normal tissue was more limited. We generated a novel antibody-drug conjugate (ADC) enfortumab vedotin comprising the human anti-nectin-4 antibody conjugated to the highly potent microtubule-disrupting agent MMAE. Hybridoma (AGS-22M6E) and CHO (ASG-22CE) versions of enfortumab vedotin (also known as ASG-22ME) ADC were able to bind to cell surface-expressed nectin-4 with high affinity and induced cell death in vitro in a dose-dependent manner. Treatment of mouse xenograft models of human breast, bladder, pancreatic, and lung cancers with enfortumab vedotin significantly inhibited the growth of all four tumor types and resulted in tumor regression of breast and bladder xenografts. Overall, these findings validate nectin-4 as an attractive therapeutic target in multiple solid tumors and support further clinical development, investigation, and application of nectin-4-targeting ADCs. Cancer Res; 76(10); 3003-13. ©2016 AACR.


Cancer Research | 2017

Abstract 2709: Evaluation of CD37 expression and binding of AGS67E, an antibody-drug conjugate (ADC) against CD37, on white blood cells (WBCs) collected from phase I non-Hodgkin lymphoma (NHL) patients

Sher Karki; Hector Aviña; Jacqueline Lackey; Ahmed Sawas; Kerry J. Savage; Raymond P. Perez; Ranjana H. Advani; Jasmine Zain; Owen A. O'Connor; Sara Gulesserian; Hui Zhao; Karen Jane Meyrick Morrison; Leonard Reyno; Fernando Doñate

AGS67E is an antibody drug conjugate (ADC) against CD37 conjugated to monomethyl auristatin E (MMAE). CD37 is expressed on normal WBCs, but is also highly expressed in NHL, CLL and AML (Pereira et al., 2015). A phase I study is currently evaluating the safety, PK and anti-cancer activity of AGS67E with or without growth factor (GF) in subjects with relapsed/refractory NHL. To assess CD37 expression on WBCs, binding of AGS67E, and potential pharmacodynamic effects, samples from subjects were collected at pre-dose, D2, D8, and D15 and analyzed by flow cytometry. CD37 expression on subject tumor samples was also evaluated by immunohistochemistry (IHC). Our results demonstrated that CD37 was highly expressed in tumor samples and that AGS67E binds to WBCs causing down-regulation of CD37, achieving saturation of binding at 24 hours post-treatment (earliest time measured) at or above 0.9 mg/kg. A dose-dependent decrease in the number of all cell types examined was observed with a nadir occurring at D8, with partial or full recovery at D15, except for neutrophils. NK and T cell counts appeared to be least impacted while neutrophils were most affected. B cell counts were extremely low pre-dose for some patients, presumably from prior therapies. In patients treated at 0.9 mg/kg and higher without GF, recovery of neutrophils was delayed beyond D15. At doses of 1.2 mg/kg and higher, use of GF resulted in a significant recovery of neutrophils by D15. The extent of cell count decreases did not correlate to the proportion of cells expressing CD37. For example, decreases in NK cells, monocytes, and, in some cases, T cells, were much greater than the proportion of cells expressing CD37. Furthermore, mature WBCs are unlikely to be affected by AGS67E. This raises the possibility that the main effect of AGS67E may be on rapidly growing precursor cells and that cells with low, or no, CD37 expression may be impacted by the membrane permeable MMAE through a by-stander effect. The effect of AGS67E on neutrophils was investigated in an in vitro assay where hematopoietic stem cells were differentiated into neutrophils. Using this method, we showed that when AGS67C antibody was conjugated to a non-cleavable, membrane impermeable auristatin (mcMMAF) less cytotoxicity to differentiating neutrophils was observed compared to AGS67E. Previously, we have shown that neutrophils secrete proteases that can liberate MMAE from ADCs (Zhao et al, 2016). These results suggest that AGS67E contributes to neutropenia through a by-stander effect, in addition to the CD37-mediated internalization of the ADC. In conclusion, the results showed that AGS67E bound to its target CD37, modulated its expression, achieved saturation of binding at doses at or above 0.9 mg/kg, and reversibly depleted WBCs, with the exception of neutrophils for which GF administration appeared to significantly improve recovery rate. Citation Format: Sher Karki, Hector Avina, Jacqueline Lackey, Ahmed Sawas, Kerry J. Savage, Raymond Perez, Ranjana Advani, Jasmine Zain, Owen A. O9Connor, Sara Gulesserian, Hui Zhao, Peng Yang, Karen Morrison, Leonard Reyno, Fernando Donate. Evaluation of CD37 expression and binding of AGS67E, an antibody-drug conjugate (ADC) against CD37, on white blood cells (WBCs) collected from phase I non-Hodgkin lymphoma (NHL) patients [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 2709. doi:10.1158/1538-7445.AM2017-2709


Cancer Research | 2016

Abstract 574: AGS62P1, a novel site-specific antibody drug conjugate targeting FLT3 exhibits potent anti-tumor activity regardless of FLT3 kinase activation status

Nandini Rudra-Ganguly; Pia M. Challita-Eid; Christine Lowe; Mike Mattie; Sung-Ju Moon; Brian A. Mendelsohn; Monica Leavitt; Cyrus Virata; Alla Verlinsky; Linnette Capo; Mi Sook Chang; Deanna L. Russell; Baljinder Randhawa; Gao Liu; René Hubert; Mary Brodey; Hector Aviña; Chunying Zhang; Joseph D. Abad; Banmeet Anand; Sher Karki; Zili An; Roland Luethy; Fernando Doñate; Daniel S. Pereira; Kendall Morrison; Ingrid B.J.K. Joseph; David R. Stover

FLT3 is a member of the class III receptor tyrosine kinase family that includes C-KIT, C-FMS and platelet derived growth factor receptor (PDGFR). FLT3 is primarily expressed in early myeloid and lymphoid progenitors and plays an important role in their proliferation and differentiation. In human leukemia, FLT3 is expressed on 70-90% acute myeloid leukemia (AML) and most B-acute lymphoblastic leukemia (B-ALL). FLT3 genetic aberrations are commonly detected in patients with AML. The most common aberration is internal tandem duplication (ITD), which occurs in 25-30% of AML patients and causes constitutive activation of FLT3. Point mutation in codon D835 of the FLT3 tyrosine kinase domain is reported in 7-10% of AML patients and also causes constitutive activation of the receptor. FLT3 small molecule inhibitors targeting the kinase domain are predominantly active against FLT3 activated AML. The restricted normal tissue expression profile and higher differential in leukemic specimens makes FLT3 amenable to antibody-based therapeutics, requiring only target expression independent of kinase activation status. Therefore, development of an antibody-drug conjugate (ADC) may provide a therapeutic alternative for AML patients. Here, we report the development of the first FLT3specific ADC, AGS62P1, employing site-specific conjugation using the non-natural amino acid, p-acetyl phenylalanine (pAF). AGS62P1 comprises a human gamma one antibody including an inserted pAF residue in each of the heavy chains. The antibody was conjugated to a potent cytotoxic payload via an oxime bond at the pAF sites, thus creating a nearly homogeneous drug distribution, with approximately 2 drug molecules per antibody. Strong binding affinity (0.1-0.9 nM) and potent in vitro cytotoxic activity (IC50 = 0.2-12 nM) was achieved in AML cell lines. The anti-FLT3 ADC was highly efficacious in AML tumor xenografts, leading to statistically significant tumor growth inhibition of both FLT3 ITD and non-ITD models. Additional characterization of both the antibody and ADC was performed, including ligand receptor interaction, degradation, internalization, and apoptosis. In summary, we have developed a site-specific ADC targeting FLT3 that exhibits potent anti-tumor activity in xenograft models regardless of FLT3 activation status. This drug can potentially offer a new and more versatile approach in targeting FLT3-expressing leukemia through a mechanism independent of FLT3 genetic aberration. Citation Format: Nandini Rudra-Ganguly, Pia M. Challita-Eid, Christine Lowe, Mike Mattie, Sung-Ju Moon, Brian A. Mendelsohn, Monica Leavitt, Cyrus Virata, Alla Verlinsky, Linnette Capo, Mi Sook Chang, Deanna L. Russell, Baljinder Randhawa, Gao Liu, Rene Hubert, Mary Brodey, Hector Avina, Chunying Zhang, Joseph D. Abad, Banmeet Anand, Sher Karki, Zili An, Roland Luethy, Fernando Donate, Daniel S. Pereira, Kendall Morrison, Ingrid B.J. Joseph, David R. Stover. AGS62P1, a novel site-specific antibody drug conjugate targeting FLT3 exhibits potent anti-tumor activity regardless of FLT3 kinase activation status. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 574.


Blood | 2005

A Humanized Anti-CD30 Monoclonal Antibody, XmAb™2513, with Enhanced In Vitro Potency Against CD30-Positive Lymphomas Mediated by High Affinity Fc-Receptor Binding.

Philip W. Hammond; Omid Vafa; Jonathan Jacinto; Jost Vielmetter; Sher Karki; Sean J. Yoder; Greg A. Lazar; Dinesh Raturi; Mike Mollet; Marie Zhu; Lori Kunkel; David F. Carmichael


Blood | 2015

AGS62P1, a Novel Anti-FLT3 Antibody Drug Conjugate, Employing Site Specific Conjugation, Demonstrates Preclinical Anti-Tumor Efficacy in AML Tumor and Patient Derived Xenografts

Nandini Rudra-Ganguly; Christine Lowe; Cyrus Virata; Monica Leavitt; Liqing Jin; Brian A. Mendelsohn; Josh Snyder; Hector Aviña; Chunying Zhang; Deanna L. Russell; Michael Mattie; Baljinder Randhawa; Gao Liu; Faisal Malik; Misty Vest; Joseph D. Abad; Christopher Kemball; René Hubert; Sher Karki; Banmeet Anand; Zili An; Jeanette Grant; John E. Dick; Fernando Doñate; Kendall Morrison; Pia M. Challita-Eid; Ingrid B.J.K. Joseph; Daniel S. Pereira; David R. Stover


Blood | 2006

Fc Engineered Anti-CD19 Monoclonal Antibodies with Enhanced In Vitro Efficacy Against Multiple Lymphoma Cell Lines.

Eugene A. Zhukovsky; Seung Y. Chu; Matthew J. Bernett; Philip W. Hammond; Sher Karki; Wei Dang; Collin Edler; Nephi Polder; Cheryl Chan; Jonathan Jacinto; John R. Desjarlais


Blood | 2004

Improving the Efficacy of Rituxan by Rational Protein Engineering.

Greg A. Lazar; Wei Dang; Omid Vafa; Sher Karki; Jost Vielmetter; Stephen Doberstein; David F. Carmichael; Robert J. Hayes


Archive | 2003

Variantes de Fc optimizadas y métodos para su generación

Gregory Alan Lazar; Arthur J. Chirino; Wei Dang; John R. Desjarlais; Stephen Kohn Doberstein; Robert J. Hayes; Sher Karki; Omid Vafa

Collaboration


Dive into the Sher Karki's collaboration.

Top Co-Authors

Avatar

John R. Desjarlais

Pennsylvania State University

View shared research outputs
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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge