Network


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

Hotspot


Dive into the research topics where Nirav Malani is active.

Publication


Featured researches published by Nirav Malani.


Nature | 2011

In vivo genome editing restores haemostasis in a mouse model of haemophilia

Hojun Li; Virginia Haurigot; Yannick Doyon; Tianjian Li; Sunnie Y. Wong; Anand S. Bhagwat; Nirav Malani; Xavier M. Anguela; Rajiv Sharma; Lacramiora Ivanciu; Samuel L. Murphy; Jonathan D. Finn; Fayaz R. Khazi; Shangzhen Zhou; David Paschon; Edward J. Rebar; Frederic D. Bushman; Philip D. Gregory; Michael C. Holmes; Katherine A. High

Editing of the human genome to correct disease-causing mutations is a promising approach for the treatment of genetic disorders. Genome editing improves on simple gene-replacement strategies by effecting in situ correction of a mutant gene, thus restoring normal gene function under the control of endogenous regulatory elements and reducing risks associated with random insertion into the genome. Gene-specific targeting has historically been limited to mouse embryonic stem cells. The development of zinc finger nucleases (ZFNs) has permitted efficient genome editing in transformed and primary cells that were previously thought to be intractable to such genetic manipulation. In vitro, ZFNs have been shown to promote efficient genome editing via homology-directed repair by inducing a site-specific double-strand break (DSB) at a target locus, but it is unclear whether ZFNs can induce DSBs and stimulate genome editing at a clinically meaningful level in vivo. Here we show that ZFNs are able to induce DSBs efficiently when delivered directly to mouse liver and that, when co-delivered with an appropriately designed gene-targeting vector, they can stimulate gene replacement through both homology-directed and homology-independent targeted gene insertion at the ZFN-specified locus. The level of gene targeting achieved was sufficient to correct the prolonged clotting times in a mouse model of haemophilia B, and remained persistent after induced liver regeneration. Thus, ZFN-driven gene correction can be achieved in vivo, raising the possibility of genome editing as a viable strategy for the treatment of genetic disease.


PLOS Pathogens | 2009

Host Cell Factors in HIV Replication: Meta-Analysis of Genome-Wide Studies

Frederic D. Bushman; Nirav Malani; Jason Fernandes; Iván D'Orso; Gerard Cagney; Tracy L. Diamond; Honglin Zhou; Daria J. Hazuda; Amy S. Espeseth; Renate König; Sourav Bandyopadhyay; Trey Ideker; Stephen P. Goff; Nevan J. Krogan; Alan D. Frankel; John A. T. Young; Sumit K. Chanda

We have analyzed host cell genes linked to HIV replication that were identified in nine genome-wide studies, including three independent siRNA screens. Overlaps among the siRNA screens were very modest (<7% for any pairwise combination), and similarly, only modest overlaps were seen in pairwise comparisons with other types of genome-wide studies. Combining all genes from the genome-wide studies together with genes reported in the literature to affect HIV yields 2,410 protein-coding genes, or fully 9.5% of all human genes (though of course some of these are false positive calls). Here we report an “encyclopedia” of all overlaps between studies (available at http://www.hostpathogen.org), which yielded a more extensively corroborated set of host factors assisting HIV replication. We used these genes to calculate refined networks that specify cellular subsystems recruited by HIV to assist in replication, and present additional analysis specifying host cell genes that are attractive as potential therapeutic targets.


The New England Journal of Medicine | 2014

A Modified γ-Retrovirus Vector for X-Linked Severe Combined Immunodeficiency

Salima Hacein-Bey-Abina; Sung-Yun Pai; H. Bobby Gaspar; Myriam Armant; Charles C. Berry; Stéphane Blanche; Jack Bleesing; Johanna Blondeau; Helen de Boer; Karen Buckland; Laure Caccavelli; Guilhem Cros; Satir De Oliveira; Karen S. Fernández; Dongjing Guo; Chad E. Harris; Gregory Hopkins; Leslie Lehmann; Annick Lim; Wendy B. London; Johannes C.M. van der Loo; Nirav Malani; Frances Male; Punam Malik; M. Angélica Marinovic; Anne Marie McNicol; Despina Moshous; Bénédicte Neven; Matías Oleastro; Capucine Picard

BACKGROUND In previous clinical trials involving children with X-linked severe combined immunodeficiency (SCID-X1), a Moloney murine leukemia virus-based γ-retrovirus vector expressing interleukin-2 receptor γ-chain (γc) complementary DNA successfully restored immunity in most patients but resulted in vector-induced leukemia through enhancer-mediated mutagenesis in 25% of patients. We assessed the efficacy and safety of a self-inactivating retrovirus for the treatment of SCID-X1. METHODS We enrolled nine boys with SCID-X1 in parallel trials in Europe and the United States to evaluate treatment with a self-inactivating (SIN) γ-retrovirus vector containing deletions in viral enhancer sequences expressing γc (SIN-γc). RESULTS All patients received bone marrow-derived CD34+ cells transduced with the SIN-γc vector, without preparative conditioning. After 12.1 to 38.7 months of follow-up, eight of the nine children were still alive. One patient died from an overwhelming adenoviral infection before reconstitution with genetically modified T cells. Of the remaining eight patients, seven had recovery of peripheral-blood T cells that were functional and led to resolution of infections. The patients remained healthy thereafter. The kinetics of CD3+ T-cell recovery was not significantly different from that observed in previous trials. Assessment of insertion sites in peripheral blood from patients in the current trial as compared with those in previous trials revealed significantly less clustering of insertion sites within LMO2, MECOM, and other lymphoid proto-oncogenes in our patients. CONCLUSIONS This modified γ-retrovirus vector was found to retain efficacy in the treatment of SCID-X1. The long-term effect of this therapy on leukemogenesis remains unknown. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01410019, NCT01175239, and NCT01129544.).


Blood | 2011

The host genomic environment of the provirus determines the abundance of HTLV-1-infected T-cell clones

Nicolas Gillet; Nirav Malani; Anat Melamed; Niall Anthony Gormley; Richard J. Carter; David R. Bentley; Charles C. Berry; Frederic D. Bushman; Graham P. Taylor; Charles R. M. Bangham

Human T-lymphotropic virus type 1 (HTLV-1) persists by driving clonal proliferation of infected T lymphocytes. A high proviral load predisposes to HTLV-1-associated diseases. Yet the reasons for the variation within and between persons in the abundance of HTLV-1-infected clones remain unknown. We devised a high-throughput protocol to map the genomic location and quantify the abundance of > 91,000 unique insertion sites of the provirus from 61 HTLV-1(+) persons and > 2100 sites from in vitro infection. We show that a typical HTLV-1-infected host carries between 500 and 5000 unique insertion sites. We demonstrate that negative selection dominates during chronic infection, favoring establishment of proviruses integrated in transcriptionally silenced DNA: this selection is significantly stronger in asymptomatic carriers. We define a parameter, the oligoclonality index, to quantify clonality. The high proviral load characteristic of HTLV-1-associated inflammatory disease results from a larger number of unique insertion sites than in asymptomatic carriers and not, as previously thought, from a difference in clonality. The abundance of established HTLV-1 clones is determined by genomic features of the host DNA flanking the provirus. HTLV-1 clonal expansion in vivo is favored by orientation of the provirus in the same sense as the nearest host gene.


PLOS Pathogens | 2011

HIV Integration Targeting: A Pathway Involving Transportin-3 and the Nuclear Pore Protein RanBP2

Karen E. Ocwieja; Troy Brady; Keshet Ronen; Alyssa Huegel; Shoshannah L. Roth; Torsten Schaller; Leo C. James; Greg J. Towers; John A. T. Young; Sumit K. Chanda; Renate König; Nirav Malani; Charles C. Berry; Frederic D. Bushman

Genome-wide siRNA screens have identified host cell factors important for efficient HIV infection, among which are nuclear pore proteins such as RanBP2/Nup358 and the karyopherin Transportin-3/TNPO3. Analysis of the roles of these proteins in the HIV replication cycle suggested that correct trafficking through the pore may facilitate the subsequent integration step. Here we present data for coupling between these steps by demonstrating that depletion of Transportin-3 or RanBP2 altered the terminal step in early HIV replication, the selection of chromosomal sites for integration. We found that depletion of Transportin-3 and RanBP2 altered integration targeting for HIV. These knockdowns reduced HIV integration frequency in gene-dense regions and near gene-associated features, a pattern that differed from that reported for depletion of the HIV integrase binding cofactor Psip1/Ledgf/p75. MLV integration was not affected by the Transportin-3 knockdown. Using siRNA knockdowns and integration targeting analysis, we also implicated several additional nuclear proteins in proper target site selection. To map viral determinants of integration targeting, we analyzed a chimeric HIV derivative containing MLV gag, and found that the gag replacement phenocopied the Transportin-3 and RanBP2 knockdowns. Thus, our data support a model in which Gag-dependent engagement of the proper transport and nuclear pore machinery mediate trafficking of HIV complexes to sites of integration.


JAMA | 2015

Outcomes Following Gene Therapy in Patients With Severe Wiskott-Aldrich Syndrome

Salima Hacein-Bey Abina; H. Bobby Gaspar; Johanna Blondeau; Laure Caccavelli; Sabine Charrier; Karen Buckland; Capucine Picard; Emmanuelle Six; Nourredine Himoudi; Kimberly Gilmour; Anne-Marie McNicol; Havinder Hara; Jinhua Xu-Bayford; Christine Rivat; Fabien Touzot; Fulvio Mavilio; Annick Lim; Jean-Marc Tréluyer; Sébastien Héritier; François Lefrere; Isabelle Pengue-Koyi; Géraldine Honnet; Stéphane Blanche; Eric Sherman; Frances Male; Charles C. Berry; Nirav Malani; Frederic D. Bushman; Alain Fischer; Aj Thrasher

IMPORTANCE Wiskott-Aldrich syndrome is a rare primary immunodeficiency associated with severe microthrombocytopenia. Partially HLA antigen-matched allogeneic hematopoietic stem cell (HSC) transplantation is often curative but is associated with significant comorbidity. OBJECTIVE To assess the outcomes and safety of autologous HSC gene therapy in Wiskott-Aldrich syndrome. DESIGN, SETTING, AND PARTICIPANTS Gene-corrected autologous HSCs were infused in 7 consecutive patients with severe Wiskott-Aldrich syndrome lacking HLA antigen-matched related or unrelated HSC donors (age range, 0.8-15.5 years; mean, 7 years) following myeloablative conditioning. Patients were enrolled in France and England and treated between December 2010 and January 2014. Follow-up of patients in this intermediate analysis ranged from 9 to 42 months. INTERVENTION A single infusion of gene-modified CD34+ cells with an advanced lentiviral vector. MAIN OUTCOMES AND MEASURES Primary outcomes were improvement at 24 months in eczema, frequency and severity of infections, bleeding tendency, and autoimmunity and reduction in disease-related days of hospitalization. Secondary outcomes were improvement in immunological and hematological characteristics and evidence of safety through vector integration analysis. RESULTS Six of the 7 patients were alive at the time of last follow-up (mean and median follow-up, 28 months and 27 months, respectively) and showed sustained clinical benefit. One patient died 7 months after treatment of preexisting drug-resistant herpes virus infection. Eczema and susceptibility to infections resolved in all 6 patients. Autoimmunity improved in 5 of 5 patients. No severe bleeding episodes were recorded after treatment, and at last follow-up, all 6 surviving patients were free of blood product support and thrombopoietic agonists. Hospitalization days were reduced from a median of 25 days during the 2 years before treatment to a median of 0 days during the 2 years after treatment. All 6 surviving patients exhibited high-level, stable engraftment of functionally corrected lymphoid cells. The degree of myeloid cell engraftment and of platelet reconstitution correlated with the dose of gene-corrected cells administered. No evidence of vector-related toxicity was observed clinically or by molecular analysis. CONCLUSIONS AND RELEVANCE This study demonstrated the feasibility of the use of gene therapy in patients with Wiskott-Aldrich syndrome. Controlled trials with larger numbers of patients are necessary to assess long-term outcomes and safety.


Molecular Therapy | 2010

LEDGF Hybrids Efficiently Retarget Lentiviral Integration Into Heterochromatin

Rik Gijsbers; Keshet Ronen; Sofie Vets; Nirav Malani; Jan De Rijck; Melissa McNeely; Frederic D. Bushman; Zeger Debyser

Correction of genetic diseases requires integration of the therapeutic gene copy into the genome of patient cells. Retroviruses are commonly used as delivery vehicles because of their precise integration mechanism, but their use has led to adverse events in which vector integration activated proto-oncogenes and contributed to leukemogenesis. Here, we show that integration by lentiviral vectors can be targeted away from genes using an artificial tethering factor. During normal lentivirus infection, the host cell-encoded transcriptional coactivator lens epithelium-derived growth factor/p75 (LEDGF/p75) binds lentiviral integrase (IN), thereby targeting integration to active transcription units and increasing the efficiency of infection. We replaced the LEDGF/p75 chromatin interaction-binding domain with CBX1. CBX1 binds histone H3 di- or trimethylated on K9, which is associated with pericentric heterochromatin and intergenic regions. The chimeric protein supported efficient transduction of lentiviral vectors and directed the integration outside of genes, near bound CBX1. Despite integration in regions rich in epigenetic marks associated with gene silencing, lentiviral vector expression remained efficient. Thus, engineered LEDGF/p75 chimeras provide technology for controlling integration site selection by lentiviral vectors.


Blood | 2011

Assessing the potential for AAV vector genotoxicity in a murine model

Hojun Li; Nirav Malani; Shari Hamilton; Alexander Schlachterman; Giulio Bussadori; Shyrie Edmonson; Rachel Shah; Valder R. Arruda; Federico Mingozzi; J. Fraser Wright; Frederic D. Bushman; Katherine A. High

Gene transfer using adeno-associated virus (AAV) vectors has great potential for treating human disease. Recently, questions have arisen about the safety of AAV vectors, specifically, whether integration of vector DNA in transduced cell genomes promotes tumor formation. This study addresses these questions with high-dose liver-directed AAV-mediated gene transfer in the adult mouse as a model (80 AAV-injected mice and 52 controls). After 18 months of follow-up, AAV-injected mice did not show a significantly higher rate of hepatocellular carcinoma compared with controls. Tumors in mice treated with AAV vectors did not have significantly different amounts of vector DNA compared with adjacent normal tissue. A novel high-throughput method for identifying AAV vector integration sites was developed and used to clone 1029 integrants. Integration patterns in tumor tissue and adjacent normal tissue were similar to each other, showing preferences for active genes, cytosine-phosphate-guanosine islands, and guanosine/cytosine-rich regions. [corrected] Gene expression data showed that genes near integration sites did not show significant changes in expression patterns compared with genes more distal to integration sites. No integration events were identified as causing increased oncogene expression. Thus, we did not find evidence that AAV vectors cause insertional activation of oncogenes and subsequent tumor formation.


Proceedings of the National Academy of Sciences of the United States of America | 2013

BET proteins promote efficient murine leukemia virus integration at transcription start sites

Amit Sharma; Ross C. Larue; Matthew R. Plumb; Nirav Malani; Frances Male; Alison Slaughter; Jacques J. Kessl; Nikolozi Shkriabai; Elizabeth Coward; Sriram Aiyer; Patrick L. Green; Li Wu; Monica J. Roth; Frederic D. Bushman; Mamuka Kvaratskhelia

The selection of chromosomal targets for retroviral integration varies markedly, tracking with the genus of the retrovirus, suggestive of targeting by binding to cellular factors. γ-Retroviral murine leukemia virus (MLV) DNA integration into the host genome is favored at transcription start sites, but the underlying mechanism for this preference is unknown. Here, we have identified bromodomain and extraterminal domain (BET) proteins (Brd2, -3, -4) as cellular-binding partners of MLV integrase. We show that purified recombinant Brd4(1-720) binds with high affinity to MLV integrase and stimulates correct concerted integration in vitro. JQ-1, a small molecule that selectively inhibits interactions of BET proteins with modified histone sites impaired MLV but not HIV-1 integration in infected cells. Comparison of the distribution of BET protein-binding sites analyzed using ChIP-Seq data and MLV-integration sites revealed significant positive correlations. Antagonism of BET proteins, via JQ-1 treatment or RNA interference, reduced MLV-integration frequencies at transcription start sites. These findings elucidate the importance of BET proteins for MLV integration efficiency and targeting and provide a route to developing safer MLV-based vectors for human gene therapy.


Blood | 2010

Dynamics of gene-modified progenitor cells analyzed by tracking retroviral integration sites in a human SCID-X1 gene therapy trial

Gary P. Wang; Charles C. Berry; Nirav Malani; Philippe Leboulch; Alain Fischer; Salima Hacein-Bey-Abina; Marina Cavazzana-Calvo; Frederic D. Bushman

X-linked severe-combined immunodeficiency (SCID-X1) has been treated by therapeutic gene transfer using gammaretroviral vectors, but insertional activation of proto-oncogenes contributed to leukemia in some patients. Here we report a longitudinal study of gene-corrected progenitor cell populations from 8 patients using 454 pyrosequencing to map vector integration sites, and extensive resampling to allow quantification of clonal abundance. The number of transduced cells infused into patients initially predicted the subsequent diversity of circulating cells. A capture-recapture analysis was used to estimate the size of the gene-corrected cell pool, revealing that less than 1/100th of the infused cells had long-term repopulating activity. Integration sites were clustered even at early time points, often near genes involved in growth control, and several patients harbored expanded cell clones with vectors integrated near the cancer-implicated genes CCND2 and HMGA2, but remain healthy. Integration site tracking also documented that chemotherapy for adverse events resulted in successful control. The longitudinal analysis emphasizes that key features of transduced cell populations--including diversity, integration site clustering, and expansion of some clones--were established early after transplantation. The approaches to sequencing and bioinformatics analysis reported here should be widely useful in assessing the outcome of gene therapy trials.

Collaboration


Dive into the Nirav Malani's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Troy Brady

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Rik Gijsbers

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Frances Male

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Keshet Ronen

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Sofie Vets

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Zeger Debyser

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jan De Rijck

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge