Network


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

Hotspot


Dive into the research topics where Abdul Qader Sukkurwala is active.

Publication


Featured researches published by Abdul Qader Sukkurwala.


Science | 2011

Autophagy-Dependent Anticancer Immune Responses Induced by Chemotherapeutic Agents in Mice

Mickaël Michaud; Isabelle Martins; Abdul Qader Sukkurwala; Sandy Adjemian; Yuting Ma; Patrizia Pellegatti; Shensi Shen; Oliver Kepp; Marie Scoazec; Grégoire Mignot; Santiago Rello-Varona; Laurie Menger; Erika Vacchelli; Lorenzo Galluzzi; François Ghiringhelli; Francesco Di Virgilio; Laurence Zitvogel; Guido Kroemer

The release of adenosine triphosphate through autophagy can promote antitumor immune responses. Antineoplastic chemotherapies are particularly efficient when they elicit immunogenic cell death, thus provoking an anticancer immune response. Here we demonstrate that autophagy, which is often disabled in cancer, is dispensable for chemotherapy-induced cell death but required for its immunogenicity. In response to chemotherapy, autophagy-competent, but not autophagy-deficient, cancers attracted dendritic cells and T lymphocytes into the tumor bed. Suppression of autophagy inhibited the release of adenosine triphosphate (ATP) from dying tumor cells. Conversely, inhibition of extracellular ATP-degrading enzymes increased pericellular ATP in autophagy-deficient tumors, reestablished the recruitment of immune cells, and restored chemotherapeutic responses but only in immunocompetent hosts. Thus, autophagy is essential for the immunogenic release of ATP from dying cells, and increased extracellular ATP concentrations improve the efficacy of antineoplastic chemotherapies when autophagy is disabled.


Immunity | 2013

Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells

Yuting Ma; Sandy Adjemian; Stephen R. Mattarollo; Takahiro Yamazaki; Laetitia Aymeric; Heng Yang; João Paulo Portela Catani; Dalil Hannani; Helene Duret; Kim Steegh; Isabelle Martins; Frederic Schlemmer; Mickaël Michaud; Oliver Kepp; Abdul Qader Sukkurwala; Laurie Menger; Erika Vacchelli; Nathalie Droin; Lorenzo Galluzzi; Roman Krzysiek; Siamon Gordon; Philip R. Taylor; Peter van Endert; Eric Solary; Mark J. Smyth; Laurence Zitvogel; Guido Kroemer

The therapeutic efficacy of anthracyclines relies on antitumor immune responses elicited by dying cancer cells. How chemotherapy-induced cell death leads to efficient antigen presentation to Txa0cells, however, remains a conundrum. We found that intratumoral CD11c(+)CD11b(+)Ly6C(hi) cells, which displayed some characteristics of inflammatory dendritic cells and included granulomonocytic precursors, were crucial for anthracycline-induced anticancer immune responses. ATP released by dying cancer cells recruited myeloid cells into tumors and stimulated the local differentiation of CD11c(+)CD11b(+)Ly6C(hi) cells. Such cells efficiently engulfed tumor antigens inxa0situ and presented them to T lymphocytes, thus vaccinating mice, upon adoptive transfer, against a challenge with cancer cells. Manipulations preventing tumor infiltration by CD11c(+)CD11b(+)Ly6C(hi) cells,xa0such as the local overexpression of ectonucleotidases, the blockade of purinergic receptors, or the neutralization of CD11b, abolished the immunexa0system-dependent antitumor activity of anthracyclines. Our results identify a subset of tumor-infiltrating leukocytes as therapy-relevant antigen-presenting cells.


OncoImmunology | 2014

Consensus guidelines for the detection of immunogenic cell death

Oliver Kepp; Laura Senovilla; Ilio Vitale; Erika Vacchelli; Sandy Adjemian; Patrizia Agostinis; Lionel Apetoh; Fernando Aranda; Vincenzo Barnaba; Norma Bloy; Laura Bracci; Karine Breckpot; David Brough; Aitziber Buqué; Maria G. Castro; Mara Cirone; María I. Colombo; Isabelle Cremer; Sandra Demaria; Luciana Dini; Aristides G. Eliopoulos; Alberto Faggioni; Silvia C. Formenti; Jitka Fucikova; Lucia Gabriele; Udo S. Gaipl; Jérôme Galon; Abhishek D. Garg; François Ghiringhelli; Nathalia A. Giese

Apoptotic cells have long been considered as intrinsically tolerogenic or unable to elicit immune responses specific for dead cell-associated antigens. However, multiple stimuli can trigger a functionally peculiar type of apoptotic demise that does not go unnoticed by the adaptive arm of the immune system, which we named “immunogenic cell death” (ICD). ICD is preceded or accompanied by the emission of a series of immunostimulatory damage-associated molecular patterns (DAMPs) in a precise spatiotemporal configuration. Several anticancer agents that have been successfully employed in the clinic for decades, including various chemotherapeutics and radiotherapy, can elicit ICD. Moreover, defects in the components that underlie the capacity of the immune system to perceive cell death as immunogenic negatively influence disease outcome among cancer patients treated with ICD inducers. Thus, ICD has profound clinical and therapeutic implications. Unfortunately, the gold-standard approach to detect ICD relies on vaccination experiments involving immunocompetent murine models and syngeneic cancer cells, an approach that is incompatible with large screening campaigns. Here, we outline strategies conceived to detect surrogate markers of ICD in vitro and to screen large chemical libraries for putative ICD inducers, based on a high-content, high-throughput platform that we recently developed. Such a platform allows for the detection of multiple DAMPs, like cell surface-exposed calreticulin, extracellular ATP and high mobility group box 1 (HMGB1), and/or the processes that underlie their emission, such as endoplasmic reticulum stress, autophagy and necrotic plasma membrane permeabilization. We surmise that this technology will facilitate the development of next-generation anticancer regimens, which kill malignant cells and simultaneously convert them into a cancer-specific therapeutic vaccine.


Cancer and Metastasis Reviews | 2011

Molecular determinants of immunogenic cell death elicited by anticancer chemotherapy

Oliver Kepp; Lorenzo Galluzzi; Isabelle Martins; Frederic Schlemmer; Sandy Adjemian; Mickaël Michaud; Abdul Qader Sukkurwala; Laurie Menger; Laurence Zitvogel; Guido Kroemer

The success of some chemo- and radiotherapeutic regimens relies on the induction of immunogenic tumor cell death and on the induction of an anticancer immune response. Cells succumbing to immunogenic cell death undergo specific changes in their surface characteristics and release pro-immunogenic factors according to a defined spatiotemporal pattern. This stimulates antigen presenting cells such as dendritic cells to efficiently take up tumor antigens, process them, and cross-prime cytotoxic T lymphocytes, thus eliciting a tumor-specific cognate immune response. Such a response can also target therapy-resistant tumor (stem) cells, thereby leading, at least in some instances, to tumor eradication. In this review, we shed some light on the molecular identity of the factors that are required for cell death to be perceived as immunogenic. We discuss the intriguing observations that the most abundant endoplasmic reticulum protein, calreticulin, the most abundant intracellular metabolite, ATP, and the most abundant non-histone chromatin-binding protein, HMGB1, can determine whether cell death is immunogenic as they appear on the surface or in the microenvironment of dying cells.


Science Translational Medicine | 2012

Cardiac glycosides exert anticancer effects by inducing immunogenic cell death.

Laurie Menger; Erika Vacchelli; Sandy Adjemian; Isabelle Martins; Yuting Ma; Shensi Shen; Takahiro Yamazaki; Abdul Qader Sukkurwala; Mickaël Michaud; Grégoire Mignot; Frederic Schlemmer; Eric Sulpice; Clara Locher; Xavier Gidrol; François Ghiringhelli; Nazanine Modjtahedi; Lorenzo Galluzzi; Fabrice Andre; Laurence Zitvogel; Oliver Kepp; Guido Kroemer

Cardiac glycosides kill cancer cells in a way that stimulates the immune response. A Cancer Double Feature—3807 A traditional chemotherapeutic drug performs a one-act play: It enters and kills a dividing cancer cell and then takes its bow. However, some chemotherapeutics have a wider range—they not only kill individual cancer cells but also do so in such a way that the dead cells function as a vaccine that primes the immune system to attack other cancer cells. Menger et al. now identify cardiac glycosides as potent inducers of this so-called immunogenic cell death. Using fluorescence microscopy to detect the hallmarks of immunogenic cell death, the authors identified cardiac glycosides, such as the heart drug digoxin, as immunogenic cell death inducers. They then verified that these drugs had anticancer effects in mice with intact immune systems but not in mice that lacked functional immunity. Cancer cells that died from digoxin exposure then effectively functioned as a vaccine—stimulating the immune system so that growth of future cancers is prevented. Indeed, human cancer patients on chemotherapy who happened to be taking the cardiac glycoside digoxin to treat other medical conditions had improved overall survival compared with patients who were not taking these drugs. Although efficacy in cancer patients remains to be formally tested, cardiac glycosides may augment chemotherapeutic response—forcing cancer to bow out. Some successful chemotherapeutics, notably anthracyclines and oxaliplatin, induce a type of cell stress and death that is immunogenic, hence converting the patient’s dying cancer cells into a vaccine that stimulates antitumor immune responses. By means of a fluorescence microscopy platform that allows for the automated detection of the biochemical hallmarks of such a peculiar cell death modality, we identified cardiac glycosides (CGs) as exceptionally efficient inducers of immunogenic cell death, an effect that was associated with the inhibition of the plasma membrane Na+- and K+-dependent adenosine triphosphatase (Na+/K+-ATPase). CGs exacerbated the antineoplastic effects of DNA-damaging agents in immunocompetent but not immunodeficient mice. Moreover, cancer cells succumbing to a combination of chemotherapy plus CGs could vaccinate syngeneic mice against a subsequent challenge with living cells of the same type. Finally, retrospective clinical analyses revealed that the administration of the CG digoxin during chemotherapy had a positive impact on overall survival in cohorts of breast, colorectal, head and neck, and hepatocellular carcinoma patients, especially when they were treated with agents other than anthracyclines and oxaliplatin.


Autophagy | 2012

Premortem autophagy determines the immunogenicity of chemotherapy-induced cancer cell death

Isabelle Martins; Mickaël Michaud; Abdul Qader Sukkurwala; Sandy Adjemian; Yuting Ma; Shensi Shen; Oliver Kepp; Laurie Menger; Erika Vacchelli; Lorenzo Galluzzi; Laurence Zitvogel; Guido Kroemer

One particular strategy to render anticancer therapies efficient consists of converting the patient’s own tumor cells into therapeutic vaccines, via the induction of immunogenic cell death (ICD). One of the hallmarks of ICD dwells in the active release of ATP by cells committed to undergo, but not yet having succumbed to, apoptosis. We observed that the knockdown of essential autophagy-related genes (ATG3, ATG5, ATG7 and BECN1) abolishes the pre-apoptotic secretion of ATP by several human and murine cancer cell lines undergoing ICD. Accordingly, autophagy-competent, but not autophagy-deficient, tumor cells treated with ICD inducers in vitro could induce a tumor-specific immune response in vivo. Cancer cell lines stably depleted of ATG5 or ATG7 normally generate tumors in vivo, and such autophagy-deficient neoplasms, upon systemic treatment with ICD inducers, exhibit the same levels of apoptosis (as monitored by nuclear shrinkage and caspase-3 activation) and necrosis (as determined by following the kinetics of HMGB1 release) as their autophagy-proficient counterparts. However, autophagy-incompetent cancers fail to release ATP, to recruit immune effectors into the tumor bed and to respond to chemotherapy in conditions in which autophagy-competent tumors do so. The intratumoral administration of ecto-ATPase inhibitors increases extracellular ATP concentrations, re-establishes the therapy-induced recruitment of dendritic cells and T cells into the tumor bed, and restores the chemotherapeutic response of autophagy-deficient cancers. Altogether, these results suggest that autophagy-incompetent tumor cells escape from chemotherapy-induced (and perhaps natural?) immunosurveillance because they are unable to release ATP.


Cytokine & Growth Factor Reviews | 2013

Crosstalk between ER stress and immunogenic cell death

Oliver Kepp; Laurie Menger; Erika Vacchelli; Clara Locher; Sandy Adjemian; Takahiro Yamazaki; Isabelle Martins; Abdul Qader Sukkurwala; Michael Michaud; Laura Senovilla; Lorenzo Galluzzi; Guido Kroemer; Laurence Zitvogel

Preclinical and clinical findings suggest that tumor-specific immune responses may be responsible--at least in part--for the clinical success of therapeutic regimens that rely on immunogenic cell death (ICD) inducers, including anthracyclines and oxaliplatin. The molecular pathways whereby some, but not all, cytotoxic agents promote bona fide ICD remain to be fully elucidated. Nevertheless, a central role for the endoplasmic reticulum (ER) stress response has been revealed in all scenarios of ICD described thus far. Hence, components of the ER stress machinery may constitute clinically relevant druggable targets for the induction of ICD. In this review, we will summarize recent findings in the field of ICD research with a special focus on ER stress mechanisms and their implication for cancer therapy.


Autophagy | 2012

Immunohistochemical detection of cytoplasmic LC3 puncta in human cancer specimens

Sylvain Ladoire; Kariman Chaba; Isabelle Martins; Abdul Qader Sukkurwala; Sandy Adjemian; Mickaël Michaud; Vichnou Poirier-Colame; Felipe Andreiuolo; Lorenzo Galluzzi; Eileen White; Mathias Rosenfeldt; Kevin M. Ryan; Laurence Zitvogel; Guido Kroemer

Autophagy is an evolutionarily conserved catabolic process that involves the entrapment of cytoplasmic components within characteristic vesicles for their delivery to and degradation within lysosomes. Alterations in autophagic signaling are found in several human diseases including cancer. Here, we describe a validated immunohistochemical protocol for the detection of LC3 puncta in human formalin-fixed, paraffin-embedded cancer specimens that can also be applied to mouse tissues. In response to systemic chemotherapy, autophagy-competent mouse tumors exhibited LC3 puncta, which did not appear in mouse cancers that had been rendered autophagy-deficient by the knockdown of Atg5 or Atg7. As compared with normal tissues, LC3 staining was moderately to highly elevated in the large majority of human cancers studied, albeit tumors of the same histological type tended to be highly heterogeneous in the number and intensity of LC3 puncta per cell. Moreover, tumor-infiltrating immune cells often were highly positive for LC3. Altogether, this protocol for LC3 staining appears suitable for the specific detection of LC3 puncta in human specimens, including tissue microarrays. We surmise that this technique can be employed for retrospective or prospective studies involving large series of human tumor samples.


OncoImmunology | 2013

ATP-dependent recruitment, survival and differentiation of dendritic cell precursors in the tumor bed after anticancer chemotherapy.

Yuting Ma; Sandy Adjemian; Heng Yang; João Paulo Portela Catani; Dalil Hannani; Isabelle Martins; Mickaël Michaud; Oliver Kepp; Abdul Qader Sukkurwala; Erika Vacchelli; Lorenzo Galluzzi; Laurence Zitvogel; Guido Kroemer

Tumor cells succumb to chemotherapy while releasing ATP. We have found that extracellular ATP attracts dendritic cell (DC) precursors into the tumor bed, facilitates their permanence in the proximity of dying cells and promotes their differentiation into mature DCs endowed with the capacity of presenting tumor-associated antigens.


OncoImmunology | 2012

Anticancer activity of cardiac glycosides: At the frontier between cell-autonomous and immunological effects.

Oliver Kepp; Laurie Menger; Erika Vacchelli; Sandy Adjemian; Isabelle Martins; Yuting Ma; Abdul Qader Sukkurwala; Mickaël Michaud; Lorenzo Galluzzi; Laurence Zitvogel; Guido Kroemer

Retrospective clinical data indicate that cardiac glycosides (CGs), notably digoxin, prolong the survival of carcinoma patients treated with conventional chemotherapy. CGs are known to influence the immune response at multiple levels. In addition, recent results suggest that CGs trigger the immunogenic demise of cancer cells, an effect that most likely contributes to their clinical anticancer activity.

Collaboration


Dive into the Abdul Qader Sukkurwala's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yuting Ma

Institut Gustave Roussy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shensi Shen

Institut Gustave Roussy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heng Yang

Institut Gustave Roussy

View shared research outputs
Researchain Logo
Decentralizing Knowledge