Network


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

Hotspot


Dive into the research topics where Kumar Sanjiv is active.

Publication


Featured researches published by Kumar Sanjiv.


Nature | 2014

MTH1 inhibition eradicates cancer by preventing sanitation of the dNTP pool.

Helge Gad; Tobias Koolmeister; Ann-Sofie Jemth; Saeed Eshtad; Sylvain A. Jacques; Cecilia E. Ström; Linda M. Svensson; Niklas Schultz; Thomas Lundbäck; Berglind O. Einarsdottir; Aljona Saleh; Camilla Göktürk; Pawel Baranczewski; Richard Svensson; Ronnie P.-A. Berntsson; Robert Gustafsson; Kia Strömberg; Kumar Sanjiv; Marie-Caroline Jacques-Cordonnier; Matthieu Desroses; Anna-Lena Gustavsson; Roger Olofsson; Fredrik Johansson; Evert Homan; Olga Loseva; Lars Bräutigam; Lars Johansson; Andreas Höglund; Anna Hagenkort; Therese Pham

Cancers have dysfunctional redox regulation resulting in reactive oxygen species production, damaging both DNA and free dNTPs. The MTH1 protein sanitizes oxidized dNTP pools to prevent incorporation of damaged bases during DNA replication. Although MTH1 is non-essential in normal cells, we show that cancer cells require MTH1 activity to avoid incorporation of oxidized dNTPs, resulting in DNA damage and cell death. We validate MTH1 as an anticancer target in vivo and describe small molecules TH287 and TH588 as first-in-class nudix hydrolase family inhibitors that potently and selectively engage and inhibit the MTH1 protein in cells. Protein co-crystal structures demonstrate that the inhibitors bind in the active site of MTH1. The inhibitors cause incorporation of oxidized dNTPs in cancer cells, leading to DNA damage, cytotoxicity and therapeutic responses in patient-derived mouse xenografts. This study exemplifies the non-oncogene addiction concept for anticancer treatment and validates MTH1 as being cancer phenotypic lethal.


Nature | 2014

Stereospecific targeting of MTH1 by ( S )-crizotinib as an anticancer strategy

Kilian Huber; E. Salah; Branka Radic; Manuela Gridling; J.M. Elkins; Alexey Stukalov; Ann-Sofie Jemth; Camilla Göktürk; Kumar Sanjiv; Kia Strömberg; Therese Pham; Ulrika Warpman Berglund; Jacques Colinge; Keiryn L. Bennett; Joanna I. Loizou; Thomas Helleday; Stefan Knapp; Giulio Superti-Furga

Activated RAS GTPase signalling is a critical driver of oncogenic transformation and malignant disease. Cellular models of RAS-dependent cancers have been used to identify experimental small molecules, such as SCH51344, but their molecular mechanism of action remains generally unknown. Here, using a chemical proteomic approach, we identify the target of SCH51344 as the human mutT homologue MTH1 (also known as NUDT1), a nucleotide pool sanitizing enzyme. Loss-of-function of MTH1 impaired growth of KRAS tumour cells, whereas MTH1 overexpression mitigated sensitivity towards SCH51344. Searching for more drug-like inhibitors, we identified the kinase inhibitor crizotinib as a nanomolar suppressor of MTH1 activity. Surprisingly, the clinically used (R)-enantiomer of the drug was inactive, whereas the (S)-enantiomer selectively inhibited MTH1 catalytic activity. Enzymatic assays, chemical proteomic profiling, kinome-wide activity surveys and MTH1 co-crystal structures of both enantiomers provide a rationale for this remarkable stereospecificity. Disruption of nucleotide pool homeostasis via MTH1 inhibition by (S)-crizotinib induced an increase in DNA single-strand breaks, activated DNA repair in human colon carcinoma cells, and effectively suppressed tumour growth in animal models. Our results propose (S)-crizotinib as an attractive chemical entity for further pre-clinical evaluation, and small-molecule inhibitors of MTH1 in general as a promising novel class of anticancer agents.


FEBS Journal | 2016

Targeting DNA repair, DNA metabolism and replication stress as anti‐cancer strategies

Jordi Carreras Puigvert; Kumar Sanjiv; Thomas Helleday

Anti‐cancer therapies targeting and damaging the DNA have been extensively used in the last 50 years since the discovery of nitrogen mustards, antimetabolites and platin agents. The use of these drugs is often limited by dose‐limiting side effects related to their poor specificity. In recent years, much effort has been put on the discovery and development of compounds that would exploit defects in DNA repair in cancer cells such as Wee1, Chk1 or PARP1 inhibitors. However, not all cancers respond to these inhibitors. Recently, new developments towards specifically targeting broader characteristics of cancer such as replication stress (RS) and lost redox homeostasis have emerged. Oncogenes induce proliferation signals, which also result in replication‐associated DNA damage, i.e. RS. Our knowledge into overall causes of RS, lesions produced and how these are signalled in cells to activate cell cycle checkpoints is evolving. Inhibition of ATR, which would normally keep non‐deleterious levels of RS, induces intolerable RS levels for cancer cells. Interestingly, links between replication and transcription appear to underlie RS along with a reduction of the dNTP pool. Remarkably, sanitization of the dNTP pool by MutT homologue 1, impeding incorporation of oxidized dNTPs into the DNA, seems to be crucial for cancer cell survival. In this minireview we present an overview of current and novel strategies to target DNA repair and exploit DNA damage to treat cancer. We present the current models for cancer‐associated RS as well as cancer phenotypic lethality. Both strategies are poised to better target cancer cells and reduce side effects.


Cell Reports | 2016

Cancer-Specific Synthetic Lethality between ATR and CHK1 Kinase Activities

Kumar Sanjiv; Anna Hagenkort; José Manuel Calderón-Montaño; Tobias Koolmeister; Philip Michael Reaper; Oliver Mortusewicz; Sylvain A. Jacques; Raoul V. Kuiper; Niklas Schultz; Martin Scobie; Peter Charlton; John Pollard; Ulrika Warpman Berglund; Mikael Altun; Thomas Helleday

Summary ATR and CHK1 maintain cancer cell survival under replication stress and inhibitors of both kinases are currently undergoing clinical trials. As ATR activity is increased after CHK1 inhibition, we hypothesized that this may indicate an increased reliance on ATR for survival. Indeed, we observe that replication stress induced by the CHK1 inhibitor AZD7762 results in replication catastrophe and apoptosis, when combined with the ATR inhibitor VE-821 specifically in cancer cells. Combined treatment with ATR and CHK1 inhibitors leads to replication fork arrest, ssDNA accumulation, replication collapse, and synergistic cell death in cancer cells in vitro and in vivo. Inhibition of CDK reversed replication stress and synthetic lethality, demonstrating that regulation of origin firing by ATR and CHK1 explains the synthetic lethality. In conclusion, this study exemplifies cancer-specific synthetic lethality between two proteins in the same pathway and raises the prospect of combining ATR and CHK1 inhibitors as promising cancer therapy.


Nature Medicine | 2017

Targeting SAMHD1 with the Vpx protein to improve cytarabine therapy for hematological malignancies

Nikolas Herold; Sean G. Rudd; Linda Ljungblad; Kumar Sanjiv; Ida Hed Myrberg; Cynthia B.J. Paulin; Yaser Heshmati; Anna Hagenkort; Juliane Kutzner; Brent D. G. Page; José Manuel Calderón-Montaño; Olga Loseva; Ann-Sofie Jemth; Lorenzo Bulli; Hanna Axelsson; Bianca Tesi; Nicholas C. K. Valerie; Andreas Höglund; Julia Bladh; Elisee Wiita; Mikael Sundin; Michael Uhlin; Georgios Rassidakis; Mats Heyman; Katja Pokrovskaja Tamm; Ulrika Warpman-Berglund; Julian Walfridsson; Sören Lehmann; Dan Grandér; Thomas Lundbäck

The cytostatic deoxycytidine analog cytarabine (ara-C) is the most active agent available against acute myelogenous leukemia (AML). Together with anthracyclines, ara-C forms the backbone of AML treatment for children and adults. In AML, both the cytotoxicity of ara-C in vitro and the clinical response to ara-C therapy are correlated with the ability of AML blasts to accumulate the active metabolite ara-C triphosphate (ara-CTP), which causes DNA damage through perturbation of DNA synthesis. Differences in expression levels of known transporters or metabolic enzymes relevant to ara-C only partially account for patient-specific differential ara-CTP accumulation in AML blasts and response to ara-C treatment. Here we demonstrate that the deoxynucleoside triphosphate (dNTP) triphosphohydrolase SAM domain and HD domain 1 (SAMHD1) promotes the detoxification of intracellular ara-CTP pools. Recombinant SAMHD1 exhibited ara-CTPase activity in vitro, and cells in which SAMHD1 expression was transiently reduced by treatment with the simian immunodeficiency virus (SIV) protein Vpx were dramatically more sensitive to ara-C-induced cytotoxicity. CRISPR–Cas9-mediated disruption of the gene encoding SAMHD1 sensitized cells to ara-C, and this sensitivity could be abrogated by ectopic expression of wild-type (WT), but not dNTPase-deficient, SAMHD1. Mouse models of AML lacking SAMHD1 were hypersensitive to ara-C, and treatment ex vivo with Vpx sensitized primary patient-derived AML blasts to ara-C. Finally, we identified SAMHD1 as a risk factor in cohorts of both pediatric and adult patients with de novo AML who received ara-C treatment. Thus, SAMHD1 expression levels dictate patient sensitivity to ara-C, providing proof-of-concept that the targeting of SAMHD1 by Vpx could be an attractive therapeutic strategy for potentiating ara-C efficacy in hematological malignancies.


Annals of Oncology | 2016

Validation and development of MTH1 inhibitors for treatment of cancer

U. Warpman Berglund; Kumar Sanjiv; Helge Gad; Christina Kalderén; Tobias Koolmeister; Therese Pham; Camilla Göktürk; Rozbeh Jafari; Gianluca Maddalo; B. Seashore-Ludlow; A. Chernobrovkin; A. Manoilov; I. S. Pateras; A. Rasti; Ann-Sofie Jemth; Ingrid Almlöf; Olga Loseva; Torkild Visnes; Berglind O. Einarsdottir; Fabienne Z. Gaugaz; Aljona Saleh; B. Platzack; Olov A. Wallner; Karl S. A. Vallin; Martin Henriksson; P. Wakchaure; S. Borhade; P. Herr; Y. Kallberg; Pawel Baranczewski

BACKGROUND Previously, we showed cancer cells rely on the MTH1 protein to prevent incorporation of otherwise deadly oxidised nucleotides into DNA and we developed MTH1 inhibitors which selectively kill cancer cells. Recently, several new and potent inhibitors of MTH1 were demonstrated to be non-toxic to cancer cells, challenging the utility of MTH1 inhibition as a target for cancer treatment. MATERIAL AND METHODS Human cancer cell lines were exposed in vitro to MTH1 inhibitors or depleted of MTH1 by siRNA or shRNA. 8-oxodG was measured by immunostaining and modified comet assay. Thermal Proteome profiling, proteomics, cellular thermal shift assays, kinase and CEREP panel were used for target engagement, mode of action and selectivity investigations of MTH1 inhibitors. Effect of MTH1 inhibition on tumour growth was explored in BRAF V600E-mutated malignant melanoma patient derived xenograft and human colon cancer SW480 and HCT116 xenograft models. RESULTS Here, we demonstrate that recently described MTH1 inhibitors, which fail to kill cancer cells, also fail to introduce the toxic oxidized nucleotides into DNA. We also describe a new MTH1 inhibitor TH1579, (Karonudib), an analogue of TH588, which is a potent, selective MTH1 inhibitor with good oral availability and demonstrates excellent pharmacokinetic and anti-cancer properties in vivo. CONCLUSION We demonstrate that in order to kill cancer cells MTH1 inhibitors must also introduce oxidized nucleotides into DNA. Furthermore, we describe TH1579 as a best-in-class MTH1 inhibitor, which we expect to be useful in order to further validate the MTH1 inhibitor concept.


Nature Communications | 2017

Synthetic lethality between androgen receptor signalling and the PARP pathway in prostate cancer

Mohammad Asim; Firas Tarish; Heather I. Zecchini; Kumar Sanjiv; Eleni Gelali; Charlie E. Massie; Ajoeb Baridi; Anne Warren; Wanfeng Zhao; Christoph Ogris; Leigh-Anne McDuffus; Patrice Mascalchi; Greg Shaw; Harveer Dev; Karan Wadhwa; Paul Wijnhoven; Josep V. Forment; Scott R. Lyons; Andy G. Lynch; Cormac O’Neill; Vincent Zecchini; Paul S. Rennie; Aria Baniahmad; Simon Tavaré; Ian G. Mills; Yaron Galanty; Nicola Crosetto; Niklas Schultz; David E. Neal; Thomas Helleday

Emerging data demonstrate homologous recombination (HR) defects in castration-resistant prostate cancers, rendering these tumours sensitive to PARP inhibition. Here we demonstrate a direct requirement for the androgen receptor (AR) to maintain HR gene expression and HR activity in prostate cancer. We show that PARP-mediated repair pathways are upregulated in prostate cancer following androgen-deprivation therapy (ADT). Furthermore, upregulation of PARP activity is essential for the survival of prostate cancer cells and we demonstrate a synthetic lethality between ADT and PARP inhibition in vivo. Our data suggest that ADT can functionally impair HR prior to the development of castration resistance and that, this potentially could be exploited therapeutically using PARP inhibitors in combination with androgen-deprivation therapy upfront in advanced or high-risk prostate cancer.Tumours with homologous recombination (HR) defects become sensitive to PARPi. Here, the authors show that androgen receptor (AR) regulates HR and AR inhibition activates the PARP pathway in vivo, thus inhibition of both AR and PARP is required for effective treatment of high risk prostate cancer.


Nature Chemical Biology | 2017

Global survey of the immunomodulatory potential of common drugs

Gregory I. Vladimer; Berend Snijder; Nikolaus Krall; Johannes W. Bigenzahn; Kilian Huber; Charles-Hugues Lardeau; Kumar Sanjiv; Anna Ringler; Ulrika Warpman Berglund; Monika Sabler; Oscar Lopez de la Fuente; Paul Knöbl; Stefan Kubicek; Thomas Helleday; Ulrich Jäger; Giulio Superti-Furga

Small molecule drugs may complement antibody-based therapies in an immune-oncology setting, yet systematic methods for the identification and characterization of the immunomodulatory properties of these entities are lacking. We surveyed the immumomodulatory potential of 1,402 small chemical molecules as defined by their ability to alter the cell-cell interactions among peripheral mononuclear leukocytes ex vivo, using automated microscopy and population-wide single-cell image analysis. Surprisingly, some 10% of the agents tested affected these cell-cell interactions differentially. The results accurately recapitulated known immunomodulatory drug classes, and revealed several clinically approved drugs that unexpectedly harbor the ability to modulate the immune system, potentially contributing to their physiological mechanism of action. For instance, the kinase inhibitor crizotinib promoted T-cell interactions with monocytes as well as with cancer cells, through inhibition of MST1R/RON (macrophage-stimulating protein receptor) and subsequent upregulation of MHC (major histocompatibility complex) expression. The approach offers an attractive platform for the personalized identification and characterization of immunomodulatory therapeutics.


Cell Cycle | 2017

SAMHD1 protects cancer cells from various nucleoside-based antimetabolites

Nikolas Herold; Sean G. Rudd; Kumar Sanjiv; Juliane Kutzner; Julia Bladh; Cynthia B.J. Paulin; Thomas Helleday; Jan-Inge Henter; Torsten Schaller

ABSTRACT Recently, we demonstrated that sterile α motif and HD domain containing protein 1 (SAMHD1) is a major barrier in acute myelogenous leukemia (AML) cells to the cytotoxicity of cytarabine (ara-C), the most important drug in AML treatment. Ara-C is intracellularly converted by the canonical dNTP synthesis pathway to ara-CTP, which serves as a substrate but not an allosteric activator of SAMHD1. Using an AML mouse model, we show here that wild type but not catalytically inactive SAMHD1 reduces ara-C treatment efficacy in vivo. Expanding the clinically relevant substrates of SAMHD1, we demonstrate that THP-1 CRISPR/Cas9 cells lacking a functional SAMHD1 gene showed increased sensitivity to the antimetabolites nelarabine, fludarabine, decitabine, vidarabine, clofarabine, and trifluridine. Within this Extra View, we discuss and build upon both these and our previously reported findings, and propose SAMHD1 is likely active against a variety of nucleoside analog antimetabolites present in anti-cancer chemotherapies. Thus, SAMHD1 may constitute a promising target to improve a wide range of therapies for both hematological and non-haematological malignancies.


Oncotarget | 2017

Glioblastoma and glioblastoma stem cells are dependent on functional MTH1

Linda Pudelko; Pegah Rouhi; Kumar Sanjiv; Helge Gad; Christina Kalderén; Andreas Höglund; Massimo Squatrito; Alberto J. Schuhmacher; Steven Edwards; Daniel Hägerstrand; Ulrika Warpman Berglund; Thomas Helleday; Lars Bräutigam

Glioblastoma multiforme (GBM) is an aggressive form of brain cancer with poor prognosis. Cancer cells are characterized by a specific redox environment that adjusts metabolism to its specific needs and allows the tumor to grow and metastasize. As a consequence, cancer cells and especially GBM cells suffer from elevated oxidative pressure which requires antioxidant-defense and other sanitation enzymes to be upregulated. MTH1, which degrades oxidized nucleotides, is one of these defense enzymes and represents a promising cancer target. We found MTH1 expression levels elevated and correlated with GBM aggressiveness and discovered that siRNA knock-down or inhibition of MTH1 with small molecules efficiently reduced viability of patient-derived GBM cultures. The effect of MTH1 loss on GBM viability was likely mediated through incorporation of oxidized nucleotides and subsequent DNA damage. We revealed that MTH1 inhibition targets GBM independent of aggressiveness as well as potently kills putative GBM stem cells in vitro. We used an orthotopic zebrafish model to confirm our results in vivo and light-sheet microscopy to follow the effect of MTH1 inhibition in GBM in real time. In conclusion, MTH1 represents a promising target for GBM therapy and MTH1 inhibitors may also be effective in patients that suffer from recurring disease.Glioblastoma multiforme (GBM) is an aggressive form of brain cancer with poor prognosis. Cancer cells are characterized by a specific redox environment that adjusts metabolism to its specific needs and allows the tumor to grow and metastasize. As a consequence, cancer cells and especially GBM cells suffer from elevated oxidative pressure which requires antioxidant-defense and other sanitation enzymes to be upregulated. MTH1, which degrades oxidized nucleotides, is one of these defense enzymes and represents a promising cancer target. We found MTH1 expression levels elevated and correlated with GBM aggressiveness and discovered that siRNA knock-down or inhibition of MTH1 with small molecules efficiently reduced viability of patient-derived GBM cultures. The effect of MTH1 loss on GBM viability was likely mediated through incorporation of oxidized nucleotides and subsequent DNA damage. We revealed that MTH1 inhibition targets GBM independent of aggressiveness as well as potently kills putative GBM stem cells in vitro. We used an orthotopic zebrafish model to confirm our results in vivo and light-sheet microscopy to follow the effect of MTH1 inhibition in GBM in real time.In conclusion, MTH1 represents a promising target for GBM therapy and MTH1 inhibitors may also be effective in patients that suffer from recurring disease.

Collaboration


Dive into the Kumar Sanjiv's collaboration.

Top Co-Authors

Avatar

Helge Gad

Karolinska Institutet

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nikolas Herold

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Juliane Kutzner

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fredrik Johansson

Royal Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Ida Hed Myrberg

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Jan-Inge Henter

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge