Network


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

Hotspot


Dive into the research topics where Matthew J. Figliola is active.

Publication


Featured researches published by Matthew J. Figliola.


Cancer Research | 2011

Reprogramming CD19-Specific T Cells with IL-21 Signaling Can Improve Adoptive Immunotherapy of B-Lineage Malignancies

Harjeet Singh; Matthew J. Figliola; Margaret J. Dawson; Helen Huls; Simon Olivares; Kirsten Switzer; Tiejuan Mi; Sourindra Maiti; Partow Kebriaei; Dean A. Lee; Richard E. Champlin; Laurence J.N. Cooper

Improving the therapeutic efficacy of T cells expressing a chimeric antigen receptor (CAR) represents an important goal in efforts to control B-cell malignancies. Recently an intrinsic strategy has been developed to modify the CAR itself to improve T-cell signaling. Here we report a second extrinsic approach based on altering the culture milieu to numerically expand CAR(+) T cells with a desired phenotype, for the addition of interleukin (IL)-21 to tissue culture improves CAR-dependent T-cell effector functions. We used electrotransfer of Sleeping Beauty system to introduce a CAR transposon and selectively propagate CAR(+) T cells on CD19(+) artificial antigen-presenting cells (aAPC). When IL-21 was present, there was preferential numeric expansion of CD19-specific T cells which lysed and produced IFN-γ in response to CD19. Populations of these numerically expanded CAR(+) T cells displayed an early memory surface phenotype characterized as CD62L(+)CD28(+) and a transcriptional profile of naïve T cells. In contrast, T cells propagated with only exogenous IL-2 tended to result in an overgrowth of CD19-specific CD4(+) T cells. Furthermore, adoptive transfer of CAR(+) T cells cultured with IL-21 exhibited improved control of CD19(+) B-cell malignancy in mice. To provide coordinated signaling to propagate CAR(+) T cells, we developed a novel mutein of IL-21 bound to the cell surface of aAPC that replaced the need for soluble IL-21. Our findings show that IL-21 can provide an extrinsic reprogramming signal to generate desired CAR(+) T cells for effective immunotherapy.


Journal of Clinical Investigation | 2016

Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells

Partow Kebriaei; Harjeet Singh; M. Helen Huls; Matthew J. Figliola; Roland L. Bassett; Simon Olivares; Bipulendu Jena; Margaret J. Dawson; Pappanaicken R. Kumaresan; Shihuang Su; Sourindra Maiti; Jianliang Dai; Branden S. Moriarity; Marie Andrée Forget; Vladimir Senyukov; Aaron Orozco; Tingting Liu; Jessica McCarty; Rineka Jackson; Judy S. Moyes; Gabriela Rondon; Muzaffar H. Qazilbash; Stefan O. Ciurea; Amin M. Alousi; Yago Nieto; Katy Rezvani; David Marin; Uday Popat; Chitra Hosing; Elizabeth J. Shpall

BACKGROUND T cells expressing antigen-specific chimeric antigen receptors (CARs) improve outcomes for CD19-expressing B cell malignancies. We evaluated a human application of T cells that were genetically modified using the Sleeping Beauty (SB) transposon/transposase system to express a CD19-specific CAR. METHODS T cells were genetically modified using DNA plasmids from the SB platform to stably express a second-generation CD19-specific CAR and selectively propagated ex vivo with activating and propagating cells (AaPCs) and cytokines. Twenty-six patients with advanced non-Hodgkin lymphoma and acute lymphoblastic leukemia safely underwent hematopoietic stem cell transplantation (HSCT) and infusion of CAR T cells as adjuvant therapy in the autologous (n = 7) or allogeneic settings (n = 19). RESULTS SB-mediated genetic transposition and stimulation resulted in 2,200- to 2,500-fold ex vivo expansion of genetically modified T cells, with 84% CAR expression, and without integration hotspots. Following autologous HSCT, the 30-month progression-free and overall survivals were 83% and 100%, respectively. After allogeneic HSCT, the respective 12-month rates were 53% and 63%. No acute or late toxicities and no exacerbation of graft-versus-host disease were observed. Despite a low antigen burden and unsupportive recipient cytokine environment, CAR T cells persisted for an average of 201 days for autologous recipients and 51 days for allogeneic recipients. CONCLUSIONS CD19-specific CAR T cells generated with SB and AaPC platforms were safe, and may provide additional cancer control as planned infusions after HSCT. These results support further clinical development of this nonviral gene therapy approach. TRIAL REGISTRATION Autologous, NCT00968760; allogeneic, NCT01497184; long-term follow-up, NCT01492036. FUNDING National Cancer Institute, private foundations, and institutional funds. Please see Acknowledgments for details.


PLOS ONE | 2013

Manufacture of Clinical-Grade CD19-Specific T Cells Stably Expressing Chimeric Antigen Receptor Using Sleeping Beauty System and Artificial Antigen Presenting Cells

Harjeet Singh; Matthew J. Figliola; Margaret J. Dawson; Simon Olivares; Ling-ling Zhang; Ge Yang; Sourindra Maiti; Pallavi R. Manuri; Vladimir Senyukov; Bipulendu Jena; Partow Kebriaei; Richard E. Champlin; Helen Huls; Laurence J.N. Cooper

Adoptive transfer of T cells expressing a CD19-specific chimeric antigen receptor (CAR) is being evaluated in multiple clinical trials. Our current approach to adoptive immunotherapy is based on a second generation CAR (designated CD19RCD28) that signals through a CD28 and CD3-ζ endodomain. T cells are electroporated with DNA plasmids from the Sleeping Beauty (SB) transposon/transposase system to express this CAR. Stable integrants of genetically modified T cells can then be retrieved when co-cultured with designer artificial antigen presenting cells (aAPC) in the presence of interleukin (IL)-2 and 21. Here, we reveal how the platform technologies of SB-mediated transposition and CAR-dependent propagation on aAPC were adapted for human application. Indeed, we have initiated clinical trials in patients with high-risk B-lineage malignancies undergoing autologous and allogeneic hematopoietic stem-cell transplantation (HSCT). We describe the process to manufacture clinical grade CD19-specific T cells derived from healthy donors. Three validation runs were completed in compliance with current good manufacturing practice for Phase I/II trials demonstrating that by 28 days of co-culture on γ-irradiated aAPC ∼1010 T cells were produced of which >95% expressed CAR. These genetically modified and propagated T cells met all quality control testing and release criteria in support of infusion.


PLOS ONE | 2010

Induction of Autophagy by Cystatin C: A Mechanism That Protects Murine Primary Cortical Neurons and Neuronal Cell Lines

Belen Tizon; Susmita Sahoo; Haung Yu; Sebastien A. Gauthier; Asok Kumar; Panaiyur S. Mohan; Matthew J. Figliola; Monika Pawlik; Anders Grubb; Yasuo Uchiyama; Urmi Bandyopadhyay; Ana Maria Cuervo; Ralph A. Nixon; Efrat Levy

Cystatin C (CysC) expression in the brain is elevated in human patients with epilepsy, in animal models of neurodegenerative conditions, and in response to injury, but whether up-regulated CysC expression is a manifestation of neurodegeneration or a cellular repair response is not understood. This study demonstrates that human CysC is neuroprotective in cultures exposed to cytotoxic challenges, including nutritional-deprivation, colchicine, staurosporine, and oxidative stress. While CysC is a cysteine protease inhibitor, cathepsin B inhibition was not required for the neuroprotective action of CysC. Cells responded to CysC by inducing fully functional autophagy via the mTOR pathway, leading to enhanced proteolytic clearance of autophagy substrates by lysosomes. Neuroprotective effects of CysC were prevented by inhibiting autophagy with beclin 1 siRNA or 3-methyladenine. Our findings show that CysC plays a protective role under conditions of neuronal challenge by inducing autophagy via mTOR inhibition and are consistent with CysC being neuroprotective in neurodegenerative diseases. Thus, modulation of CysC expression has therapeutic implications for stroke, Alzheimers disease, and other neurodegenerative disorders.


Journal of Immunotherapy | 2013

Sleeping beauty system to redirect T-cell specificity for human applications

Sourindra Maiti; Helen Huls; Harjeet Singh; Margaret J. Dawson; Matthew J. Figliola; Simon Olivares; Pullavathi Rao; Yi Jue Zhao; Asha S. Multani; Ge Yang; Ling Zhang; Denise L. Crossland; Sonny Ang; Hiroki Torikai; Brian Rabinovich; Dean A. Lee; Partow Kebriaei; Perry B. Hackett; Richard E. Champlin; Laurence J.N. Cooper

The Sleeping Beauty (SB) transposon/transposase DNA plasmid system is used to genetically modify cells for long-term transgene expression. We adapted the SB system for human application and generated T cells expressing a chimeric antigen receptor (CAR) specific for CD19. Electrotransfer of CD19-specific SB DNA plasmids in peripheral blood mononuclear cells and propagation on CD19+ artificial antigen presenting cells was used to numerically expand CD3+ T cells expressing CAR. By day 28 of coculture, >90% of expanded CD3+ T cells expressed CAR. CAR+ T cells specifically killed CD19+ target cells and consisted of subsets expressing biomarkers consistent with central memory, effector memory, and effector phenotypes. CAR+ T cells contracted numerically in the absence of the CD19 antigen, did not express SB11 transposase, and maintained a polyclonal TCR V&agr; and TCR V&bgr; repertoire. Quantitative fluorescence in situ hybridization revealed that CAR+ T cells preserved the telomere length. Quantitative polymerase chain reaction and fluorescence in situ hybridization showed CAR transposon integrated on average once per T-cell genome. CAR+ T cells in peripheral blood can be detected by quantitative polymerase chain reaction at a sensitivity of 0.01%. These findings lay the groundwork as the basis of our first-in-human clinical trials of the nonviral SB system for the investigational treatment of CD19+ B-cell malignancies (currently under 3 INDs: 14193, 14577, and 14739).


Journal of Visualized Experiments | 2013

Clinical application of Sleeping Beauty and artificial antigen presenting cells to genetically modify T cells from peripheral and umbilical cord blood.

M. Helen Huls; Matthew J. Figliola; Margaret J. Dawson; Simon Olivares; Partow Kebriaei; Elizabeth J. Shpall; Richard E. Champlin; Harjeet Singh; Laurence J.N. Cooper

The potency of clinical-grade T cells can be improved by combining gene therapy with immunotherapy to engineer a biologic product with the potential for superior (i) recognition of tumor-associated antigens (TAAs), (ii) persistence after infusion, (iii) potential for migration to tumor sites, and (iv) ability to recycle effector functions within the tumor microenvironment. Most approaches to genetic manipulation of T cells engineered for human application have used retrovirus and lentivirus for the stable expression of CAR1-3. This approach, although compliant with current good manufacturing practice (GMP), can be expensive as it relies on the manufacture and release of clinical-grade recombinant virus from a limited number of production facilities. The electro-transfer of nonviral plasmids is an appealing alternative to transduction since DNA species can be produced to clinical grade at approximately 1/10th the cost of recombinant GMP-grade virus. To improve the efficiency of integration we adapted Sleeping Beauty (SB) transposon and transposase for human application4-8. Our SB system uses two DNA plasmids that consist of a transposon coding for a gene of interest (e.g. 2nd generation CD19-specific CAR transgene, designated CD19RCD28) and a transposase (e.g. SB11) which inserts the transgene into TA dinucleotide repeats9-11. To generate clinically-sufficient numbers of genetically modified T cells we use K562-derived artificial antigen presenting cells (aAPC) (clone #4) modified to express a TAA (e.g. CD19) as well as the T cell costimulatory molecules CD86, CD137L, a membrane-bound version of interleukin (IL)-15 (peptide fused to modified IgG4 Fc region) and CD64 (Fc-γ receptor 1) for the loading of monoclonal antibodies (mAb)12. In this report, we demonstrate the procedures that can be undertaken in compliance with cGMP to generate CD19-specific CAR+ T cells suitable for human application. This was achieved by the synchronous electro-transfer of two DNA plasmids, a SB transposon (CD19RCD28) and a SB transposase (SB11) followed by retrieval of stable integrants by the every-7-day additions (stimulation cycle) of γ-irradiated aAPC (clone #4) in the presence of soluble recombinant human IL-2 and IL-2113. Typically 4 cycles (28 days of continuous culture) are undertaken to generate clinically-appealing numbers of T cells that stably express the CAR. This methodology to manufacturing clinical-grade CD19-specific T cells can be applied to T cells derived from peripheral blood (PB) or umbilical cord blood (UCB). Furthermore, this approach can be harnessed to generate T cells to diverse tumor types by pairing the specificity of the introduced CAR with expression of the TAA, recognized by the CAR, on the aAPC.


PLOS ONE | 2015

Sleeping Beauty transposition of chimeric antigen receptors targeting receptor tyrosine kinase-like orphan receptor-1 (ROR1) into diverse memory T-cell populations

Drew C. Deniger; Jianqiang Yu; M. Helen Huls; Matthew J. Figliola; Tiejuan Mi; Sourindra Maiti; George F. Widhopf; Lenka V. Hurton; Radhika Thokala; Harjeet Singh; Simon Olivares; Richard E. Champlin; William G. Wierda; Thomas J. Kipps; Laurence J.N. Cooper

T cells modified with chimeric antigen receptors (CARs) targeting CD19 demonstrated clinical activity against some B-cell malignancies. However, this is often accompanied by a loss of normal CD19+ B cells and humoral immunity. Receptor tyrosine kinase-like orphan receptor-1 (ROR1) is expressed on sub-populations of B-cell malignancies and solid tumors, but not by healthy B cells or normal post-partum tissues. Thus, adoptive transfer of T cells specific for ROR1 has potential to eliminate tumor cells and spare healthy tissues. To test this hypothesis, we developed CARs targeting ROR1 in order to generate T cells specific for malignant cells. Two Sleeping Beauty transposons were constructed with 2nd generation ROR1-specific CARs signaling through CD3ζ and either CD28 (designated ROR1RCD28) or CD137 (designated ROR1RCD137) and were introduced into T cells. We selected for T cells expressing CAR through co-culture with γ-irradiated activating and propagating cells (AaPC), which co-expressed ROR1 and co-stimulatory molecules. Numeric expansion over one month of co-culture on AaPC in presence of soluble interleukin (IL)-2 and IL-21 occurred and resulted in a diverse memory phenotype of CAR+ T cells as measured by non-enzymatic digital array (NanoString) and multi-panel flow cytometry. Such T cells produced interferon-γ and had specific cytotoxic activity against ROR1+ tumors. Moreover, such cells could eliminate ROR1+ tumor xenografts, especially T cells expressing ROR1RCD137. Clinical trials will investigate the ability of ROR1-specific CAR+ T cells to specifically eliminate tumor cells while maintaining normal B-cell repertoire.


Integrative Biology | 2013

Imaging of genetically engineered T cells by PET using gold nanoparticles complexed to Copper-64

Parijat Bhatnagar; Zheng Li; Yoonsu Choi; Jianfeng Guo; Feng Li; Daniel Y. Lee; Matthew J. Figliola; Helen Huls; Dean A. Lee; Tomasz Zal; King C. Li; Laurence J.N. Cooper

Adoptive transfer of primary T cells genetically modified to have desired specificity can exert an anti-tumor response in some patients. To improve our understanding of their therapeutic potential we have developed a clinically-appealing approach to reveal their in vivo biodistribution using nanoparticles that serve as a radiotracer for imaging by positron emission tomography (PET). T cells electroporated with DNA plasmids from the Sleeping Beauty transposon-transposase system to co-express a chimeric antigen receptor (CAR) specific for CD19 and Firefly luciferase (ffLuc) were propagated on CD19(+) K562-derived artificial antigen presenting cells. The approach to generating our clinical-grade CAR(+) T cells was adapted for electro-transfer of gold nanoparticles (GNPs) functionalized with (64)Cu(2+) using the macrocyclic chelator (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid, DOTA) and polyethyleneglycol (GNP-(64)Cu/PEG2000). MicroPET/CT was used to visualize CAR(+)EGFPffLucHyTK(+)GNP-(64)Cu/PEG2000(+) T cells and correlated with bioluminescence imaging. These data demonstrate that GNPs conjugated with (64)Cu(2+) can be prepared as a radiotracer for PET and used to image T cells using an approach that has translational implications.


Molecular Therapy | 2016

278. Next-Generation Non-Viral Gene Transfer to Redirect T-Cell Specificity

Harjeet Singh; Mary Helen Huls; Matthew J. Figliola; Ling Zhang; Tiejuan Mi; Sourindra Maiti; Gabrielle Romain; Simon Olivares; Shihuang Su; Lenka V. Hurton; Navin Varadarajan; Laurence J.N. Cooper; Partow Kebriaei

Non-viral gene transfer using the Sleeping Beauty (SB) transposon/transposase system has been successfully tested in humans to express a chimeric antigen receptor (CAR) to redirect T-cell specificity to CD19. This system has been modified to (i) improve the design of the CD19-specific CAR and (ii) reduce the time in culture to 14 days. Our previous clinical trials infused T cells expressing a 2nd generation CAR (designated CD19RCD28) with an IgG4-Fc stalk that activated via chimeric CD28 and CD3ζ. To evaluate the length of extracellular domain on function, we tested four CD19-specific CARs with two long [IgG4-Fc (CD19RCD28) and EQ (L235E and N297Q) mutant IgG4-Fc (CD19R*CD28)], medium (CD8α hinge, CD19RCD8CD28) and short (12aa IgG1 hinge, CD19R12aaCD28) stalks which all signaled through chimeric CD28 and CD3ζ endodomains. Generation of our T cells is based on electro-transfer of CARs coded by the SB system and antigen-specific stimulation through activating and K562-derived propagating cells (AaPC) in the presence of exogenous cytokines. After electro-transfer of SB-derived DNA plasmids, T cells were selectively propagated with either a new two-weekly (2x) or standard four-weekly (4x) additions of AaPC. All genetically modified T cells were capable of specific lysis of CD19+ tumor targets and producing IFN-γ in response to CD19+ stimulator cells. Serial killing was tested using massively parallel microscopy to observe single T cells and we observed that CDl9RCD8CD28+ T cells exhibited superior ability to partake in multiple killing events. CAR+ T cells were further tested in vivo for their ability to control CD19+ leukemia in a mouse model of minimal residual disease as well as established disease (Figure A and BFigure A and B). We found that T cells expressing modified CARs (CD19R*CD28, CD19RCD8CD28, CD19R12aaCD28) with reduced ability to bind to Fc gamma receptors (FcγR) were able to control leukemia more efficiently in mice compared to T cells expressing CD19RCD28. The CD19RCD8CD28 CAR was superior in controlling disease in the model of minimal residual disease compared with the CAR design evaluated in our prior clinical trials. T cells expressing CD19R*CD28 and CD19RCD8CD28 were then evaluated in 2x stimulation cycle. Both the 4x CAR+ T cells had similar CAR expression (>70%) whereas the 2x CAR+ T cells exhibited reduced CAR expression (~40%). The 2x CAR+ T cells expressed markers associated with less differentiated state of naive-like and memory T cells when compared to 4x CAR+ T cells, which was supported by measurement of mRNA species using bar-coded probes. The efficacy of the CAR+ T cells was tested in mice bearing established CD19+ leukemia and we observed superior survival in mice receiving the 2x CAR+ T cells compared with the 4x CAR+ T cells (Figure CFigure C). These data depict that length of extracellular domain and its associated binding to FcγR improves T-cell effector functions and that decreasing the time in culture can improve control of leukemia in vivo. These data support the use of cDl9RCD8CD28 testing in a next-generation clinical trial (IND# 16474).View Large Image | Download PowerPoint Slide


Blood | 2014

Adoptive Therapy Using Sleeping Beauty Gene Transfer System and Artificial Antigen Presenting Cells to Manufacture T Cells Expressing CD19-Specific Chimeric Antigen Receptor

Partow Kebriaei; Helen Huls; Harjeet Singh; Simon Olivares; Matthew J. Figliola; Sourindra Maiti; Su Shihuang; Pappanaicken R Kumar; Bipulendu Jena; Marie Andrée Forget; Sonny Ang; Jackson Rineka; Tingting Liu; Ian McNiece; Gabriela Rondon; Perry B. Hackett; William G. Wierda; Susan O'Brien; Michael J. Keating; Hagop M. Kantarjian; Chitra Hosing; Elizabeth J. Shpall; Richard E. Champlin; Laurence J.N. Cooper

Collaboration


Dive into the Matthew J. Figliola's collaboration.

Top Co-Authors

Avatar

Laurence J.N. Cooper

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Richard E. Champlin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Harjeet Singh

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Simon Olivares

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Helen Huls

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Margaret J. Dawson

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Partow Kebriaei

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sourindra Maiti

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth J. Shpall

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dean A. Lee

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge