Network


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

Hotspot


Dive into the research topics where Maksim Mamonkin is active.

Publication


Featured researches published by Maksim Mamonkin.


Journal of Clinical Investigation | 2016

Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape

Meenakshi Hegde; Malini Mukherjee; Zakaria Grada; Antonella Pignata; Daniel Landi; Shoba A. Navai; Amanda Wakefield; Kristen Fousek; Kevin Bielamowicz; Kevin Chow; Vita S. Brawley; Tiara Byrd; Simone Krebs; Stephen Gottschalk; Winfried S. Wels; Matthew L. Baker; Gianpietro Dotti; Maksim Mamonkin; Malcolm K. Brenner; Jordan S. Orange; Nabil Ahmed

In preclinical models of glioblastoma, antigen escape variants can lead to tumor recurrence after treatment with CAR T cells that are redirected to single tumor antigens. Given the heterogeneous expression of antigens on glioblastomas, we hypothesized that a bispecific CAR molecule would mitigate antigen escape and improve the antitumor activity of T cells. Here, we created a CAR that joins a HER2-binding scFv and an IL13Rα2-binding IL-13 mutein to make a tandem CAR exodomain (TanCAR) and a CD28.ζ endodomain. We determined that patient TanCAR T cells showed distinct binding to HER2 or IL13Rα2 and had the capability to lyse autologous glioblastoma. TanCAR T cells exhibited activation dynamics that were comparable to those of single CAR T cells upon encounter of HER2 or IL13Rα2. We observed that TanCARs engaged HER2 and IL13Rα2 simultaneously by inducing HER2-IL13Rα2 heterodimers, which promoted superadditive T cell activation when both antigens were encountered concurrently. TanCAR T cell activity was more sustained but not more exhaustible than that of T cells that coexpressed a HER2 CAR and an IL13Rα2 CAR, T cells with a unispecific CAR, or a pooled product. In a murine glioblastoma model, TanCAR T cells mitigated antigen escape, displayed enhanced antitumor efficacy, and improved animal survival. Thus, TanCAR T cells show therapeutic potential to improve glioblastoma control by coengaging HER2 and IL13Rα2 in an augmented, bivalent immune synapse that enhances T cell functionality and reduces antigen escape.


Blood | 2015

A T-cell-directed chimeric antigen receptor for the selective treatment of T-cell malignancies

Maksim Mamonkin; Rayne H. Rouce; Haruko Tashiro; Malcolm K. Brenner

Options for targeted therapy of T-cell malignancies remain scarce. Recent clinical trials demonstrated that chimeric antigen receptors (CARs) can effectively redirect T lymphocytes to eradicate lymphoid malignancies of B-cell origin. However, T-lineage neoplasms remain a more challenging task for CAR T cells due to shared expression of most targetable surface antigens between normal and malignant T cells, potentially leading to fratricide of CAR T cells or profound immunodeficiency. Here, we report that T cells transduced with a CAR targeting CD5, a common surface marker of normal and neoplastic T cells, undergo only limited fratricide and can be expanded long-term ex vivo. These CD5 CAR T cells effectively eliminate malignant T-cell acute lymphoblastic leukemia (T-ALL) and T-cell lymphoma lines in vitro and significantly inhibit disease progression in xenograft mouse models of T-ALL. These data support the therapeutic potential of CD5 CAR in patients with T-cell neoplasms.


Blood | 2017

CD7-edited T cells expressing a CD7-specific CAR for the therapy of T-cell malignancies

Diogo Gomes-Silva; Madhuwanti Srinivasan; Sandhya Sharma; Ciaran M Lee; Dimitrios L. Wagner; Timothy H. Davis; Rayne H. Rouce; Gang Bao; Malcolm K. Brenner; Maksim Mamonkin

Extending the success of chimeric antigen receptor (CAR) T cells to T-cell malignancies is problematic because most target antigens are shared between normal and malignant cells, leading to CAR T-cell fratricide. CD7 is a transmembrane protein highly expressed in acute T-cell leukemia (T-ALL) and in a subset of peripheral T-cell lymphomas. Normal expression of CD7 is largely confined to T cells and natural killer (NK) cells, reducing the risk of off-target-organ toxicity. Here, we show that the expression of a CD7-specific CAR impaired expansion of transduced T cells because of residual CD7 expression and the ensuing fratricide. We demonstrate that targeted genomic disruption of the CD7 gene prevented this fratricide and enabled expansion of CD7 CAR T cells without compromising their cytotoxic function. CD7 CAR T cells produced robust cytotoxicity against malignant T-cell lines and primary tumors and were protective in a mouse xenograft model of T-ALL. Although CD7 CAR T cells were also toxic against unedited (CD7+) T and NK lymphocytes, we show that the CD7-edited T cells themselves can respond to viral peptides and therefore could be protective against pathogens. Hence, genomic disruption of a target antigen overcomes fratricide of CAR T cells and establishes the feasibility of using CD7 CAR T cells for the targeted therapy of T-cell malignancies.


Journal for ImmunoTherapy of Cancer | 2015

Early transduction produces highly functional chimeric antigen receptor-modified virus-specific T-cells with central memory markers: a Production Assistant for Cell Therapy (PACT) translational application

Jiali Sun; Leslie E. Huye; Natalia Lapteva; Maksim Mamonkin; Manasa Hiregange; Brandon Ballard; Olga Dakhova; Darshana Raghavan; April G. Durett; Serena K. Perna; Bilal Omer; Lisa Rollins; Ann M. Leen; Juan F. Vera; Gianpietro Dotti; Adrian P. Gee; Malcolm K. Brenner; Douglas Myers; Cliona M. Rooney

BackgroundVirus-specific T-cells (VSTs) proliferate exponentially after adoptive transfer into hematopoietic stem cell transplant (HSCT) recipients, eliminate virus infections, then persist and provide long-term protection from viral disease. If VSTs behaved similarly when modified with tumor-specific chimeric antigen receptors (CARs), they should have potent anti-tumor activity. This theory was evaluated by Cruz et al. in a previous clinical trial with CD19.CAR-modified VSTs, but there was little apparent expansion of these cells in patients. In that study, VSTs were gene-modified on day 19 of culture and we hypothesized that by this time, sufficient T-cell differentiation may have occurred to limit the subsequent proliferative capacity of the transduced T-cells. To facilitate the clinical testing of this hypothesis in a project supported by the NHLBI-PACT mechanism, we developed and optimized a good manufacturing practices (GMP) compliant method for the early transduction of VSTs directed to Epstein-Barr virus (EBV), Adenovirus (AdV) and cytomegalovirus (CMV) using a CAR directed to the tumor-associated antigen disialoganglioside (GD2).ResultsAd-CMVpp65-transduced EBV-LCLs effectively stimulated VSTs directed to all three viruses (triVSTs). Transduction efficiency on day three was increased in the presence of cytokines and high-speed centrifugation of retroviral supernatant onto retronectin-coated plates, so that under optimal conditions up to 88% of tetramer-positive VSTs expressed the GD2.CAR. The average transduction efficiency of early-and late transduced VSTs was 55 ± 4% and 22 ± 5% respectively, and early-transduced VSTs maintained higher frequencies of T cells with central memory or intermediate memory phenotypes. Early-transduced VSTs also had higher proliferative capacity and produced higher levels of TH1 cytokines IL-2, TNF-α, IFN-γ, MIP-1α, MIP-1β and other cytokines in vitro.ConclusionsWe developed a rapid and GMP compliant method for the early transduction of multivirus-specific T-cells that allowed stable expression of high levels of a tumor directed CAR. Since a proportion of early-transduced CAR-VSTs had a central memory phenotype, they should expand and persist in vivo, simultaneously protecting against infection and targeting residual malignancy. This manufacturing strategy is currently under clinical investigation in patients receiving allogeneic HSCT for relapsed neuroblastoma and B-cell malignancies (NCT01460901 using a GD2.CAR and NCT00840853 using a CD19.CAR).


Immunology Letters | 2013

Differential roles of KLF4 in the development and differentiation of CD8+ T cells

Maksim Mamonkin; Ye Shen; Ping-Hsien Lee; Monica Puppi; Chun Shik Park; H. Daniel Lacorazza

The transcription factor Krüppel-like factor 4 (KLF4) can activate or repress gene expression in a cell-context dependent manner. We have previously shown that KLF4 inhibits the proliferation of naïve CD8(+) T cells in vitro downstream of the transcription factor ELF4. In this work, we describe a novel role of KLF4 in the differentiation of CD8(+) T cells upon infection. Loss of KLF4 had minimal effect on thymic T cell development and distribution of mature T cells in the spleen, blood, and lymph nodes. KLF4-deficient naïve CD8(+) T cells also displayed normal homeostatic proliferation upon adoptive transfer into lymphopenic hosts. However, activation of KLF4-deficient naïve CD8(+) T cells by in vitro TCR crosslink and co-stimulation resulted in increased proliferation. Furthermore, naïve KLF4-deficient OT-I CD8(+) T cells generated increased numbers of functional memory CD8(+) T cells compared to wild type OT-I CD8(+) T cells co-injected in the same recipient in both primary and recall responses to Listeria monocytogenes-OVA. Collectively, our data demonstrate that KLF4 regulates differentiation of functional memory CD8(+) T cells while sparing development and homeostasis of naïve CD8(+) T cells.


Immunology Letters | 2011

FCRL6 receptor: Expression and associated proteins

Sergey V. Kulemzin; Alina Y. Zamoshnikova; Mariya Yurchenko; Nazarii Y. Vitak; Alexander M. Najakshin; Svetlana A. Fayngerts; Nikolai A. Chikaev; Evdokiya S. Reshetnikova; Natalia M. Kashirina; Mikhail M. Peclo; P. N. Rutkevich; Alexander Y. Shevelev; Elena V. Yanushevskaya; Konstantin O. Baranov; Maksim Mamonkin; Tatjana N. Vlasik; Svetlana P. Sidorenko; Alexander V. Taranin; Ludmila V. Mechetina

FCRL6 receptor is a more recently identified representative of the FCRL family. We generated a panel of mouse mAbs to baculovirus-derived recombinant FCRL6 protein. The clone 7B2 was found to specifically recognize a 63kDa protein expressed preferentially on the surface of CD8 T and CD56 NK cells in human peripheral blood and spleen. The clone 7B2 reacts with FCRL6 in Western blotting, FACS, and immunohistochemistry. In the T cell lineage, FCRL6 functions in antigen-experienced cells. Mitogenic stimulation of PB leukocytes in vitro resulted in an abrogation of the FCRL6 gene expression. We found a significant decrease in the FCRL6 gene expression in peripheral T cells of patients with certain autoimmune and blood diseases, and its upregulation at the late stages of HIV infection. Study of the FCRL6 association with signaling molecules showed its ability to recruit SHP-1, SHP-2, SHIP-1, and SHIP-2 phosphatases, and also adaptor protein Grb2 through phosphorylated cytoplasmic tyrosines. The current results demonstrate inhibitory potential of FCRL6 and suggest its possible involvement in modulation of CTL effector functions in various immune disorders.


Cancer immunology research | 2018

Reversible Transgene Expression Reduces Fratricide and Permits 4-1BB Costimulation of CAR T Cells Directed to T-cell Malignancies

Maksim Mamonkin; Malini Mukherjee; Madhuwanti Srinivasan; Sandhya Sharma; Diogo Gomes-Silva; Feiyan Mo; Giedre Krenciute; Jordan S. Orange; Malcolm K. Brenner

CAR T cells targeting malignant T cells can form unwanted cytotoxic immunologic synapses between themselves, impairing their survival. CAR-derived 4-1BB costimulation stabilized these synapses, and reversing CAR expression overcame unwanted fratricide while retaining antitumor activity. T cells expressing second-generation chimeric antigen receptors (CARs) specific for CD5, a T-cell surface marker present on normal and malignant T cells, can selectively kill tumor cells. We aimed to improve this killing by substituting the CD28 costimulatory endodomain (28.z) with 4-1BB (BB.z), as 28.z CD5 CAR T cells rapidly differentiated into short-lived effector cells. In contrast, 4-1BB costimulation is known to promote development of the central memory subpopulation. Here, we found BB.z CD5 CAR T cells had impaired growth compared with 28.z CD5.CAR T cells, due to increased T-cell–T-cell fratricide. We demonstrate that TRAF signaling from the 4-1BB endodomain upregulated the intercellular adhesion molecule 1, which stabilized the fratricidal immunologic synapse between CD5 CAR T cells. As the surviving BB.z CD5 CAR T cells retained the desired central memory phenotype, we aimed to circumvent the 4-1BB–mediated toxicity using a regulated expression system that reversibly inhibits CAR expression. This system minimized CAR signaling and T-cell fratricide during in vitro expansion in the presence of a small-molecule inhibitor, and restored CAR expression and antitumor function of transduced T cells in vivo. These studies reveal a mechanism by which 4-1BB costimulation impairs expansion of CD5 CAR T cells and offer a solution to mitigate this toxicity. Cancer Immunol Res; 6(1); 47–58. ©2017 AACR.


European Journal of Immunology | 2014

Transcription factor ELF4 promotes development and function of memory CD8+ T cells in Listeria monocytogenes infection

Maksim Mamonkin; Monica Puppi; H. Daniel Lacorazza

Most differentiated CD8+ T cells die off at the end of an infection, revealing two main subsets of memory T cells — central and effector memory — which can be found in lymphoid tissues or circulating through nonlymphoid organs, respectively. The cell intrinsic regulation of the differentiation of CD8+ T cells to effector and central memory remains poorly studied. Herein, we describe a novel role of the ETS transcription factor ELF4 in the development and function of memory CD8+ T cells following infection with Listeria monocytogenes. Adoptively transferred Elf4−/− naïve CD8+ T cells produced lower numbers of effector memory CD8+ T cells despite a normal pool of central memory. This was caused by suboptimal priming and decreased survival of CD8+ T cells at the peak of response while enhanced Notch1 signaling and upregulation of eomesodermin correlated with “normal” development of Elf4−/− central memory. Finally, loss of ELF4 impaired the expansion of both central and effector memory CD8+ T cells in a recall response by also activating Notch1 signaling. Altogether, ELF4 emerges as a novel transcriptional regulator of CD8+ T‐cell differentiation in response to infection.


Nature | 2018

A homing system targets therapeutic T cells to brain cancer

Heba Samaha; Antonella Pignata; Kristen Fousek; Jun Ren; Fong Lam; Fabio Stossi; Julien Dubrulle; Vita S. Salsman; Shanmugarajan Krishnan; Sung-Ha Hong; Matthew L. Baker; Ankita Shree; Ahmed Z. Gad; Thomas Shum; Dai Fukumura; Tiara Byrd; Malini Mukherjee; Sean P. Marrelli; Jordan S. Orange; Sujith Joseph; Poul H. Sorensen; Michael D. Taylor; Meenakshi Hegde; Maksim Mamonkin; Rakesh K. Jain; Shahenda El-Naggar; Nabil Ahmed

Successful T cell immunotherapy for brain cancer requires that the T cells can access tumour tissues, but this has been difficult to achieve. Here we show that, in contrast to inflammatory brain diseases such as multiple sclerosis, where endothelial cells upregulate ICAM1 and VCAM1 to guide the extravasation of pro-inflammatory cells, cancer endothelium downregulates these molecules to evade immune recognition. By contrast, we found that cancer endothelium upregulates activated leukocyte cell adhesion molecule (ALCAM), which allowed us to overcome this immune-evasion mechanism by creating an ALCAM-restricted homing system (HS). We re-engineered the natural ligand of ALCAM, CD6, in a manner that triggers initial anchorage of T cells to ALCAM and conditionally mediates a secondary wave of adhesion by sensitizing T cells to low-level ICAM1 on the cancer endothelium, thereby creating the adhesion forces necessary to capture T cells from the bloodstream. Cytotoxic HS T cells robustly infiltrated brain cancers after intravenous injection and exhibited potent antitumour activity. We have therefore developed a molecule that targets the delivery of T cells to brain cancer.Therapeutic T cells bearing ligands engineered to optimize adhesion and transmigration through the blood–brain barrier can be targeted to brain tumours.


Nature Medicine | 2017

Exhausting alloreactivity of donor-derived CAR T cells

Maksim Mamonkin; Helen E. Heslop

A study in mouse models of allogeneic stem cell transplantation with donor-derived CD19 chimeric antigen receptor (CAR) T cells for the treatment of relapsed B cell malignancies indicates that T cell exhaustion might have a role in preventing allogeneic reactivity of CD19 CAR T cells.

Collaboration


Dive into the Maksim Mamonkin's collaboration.

Top Co-Authors

Avatar

Malcolm K. Brenner

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

Malini Mukherjee

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Cliona M. Rooney

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

Jordan S. Orange

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Madhuwanti Srinivasan

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

Gianpietro Dotti

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nabil Ahmed

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Olga Dakhova

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

Rayne H. Rouce

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge