Abstract
It is now clear that the immune system plays a critical role not only during oncogenesis and tumor progression, but also as established neoplastic lesions respond to therapy. Selected cytotoxic chemicals can indeed elicit immunogenic cell death, a functionally peculiar type of apoptosis that stimulates tumor-specific cognate immune responses. Such immunogenic chemotherapeutics include cyclophosphamide, doxorubicin and oxaliplatin (which are approved by FDA for the treatment of various hematological and solid malignancies), mitoxantrone (which is currently employed both as an anticancer agent and against multiple sclerosis) and patupilone (a microtubular poison in clinical development). One year ago, in the second issue of OncoImmunology, we discussed the scientific rationale behind immunogenic chemotherapy and reviewed the status of recent clinical trials investigating the off-label use of cyclophosphamide, doxorubicin, oxaliplatin and mitoxantrone in cancer patients. Here, we summarize the latest developments in this area of clinical research, covering both high-impact studies that have been published during the last 13 months and clinical trials that have been initiated in the same period to assess the antineoplastic profile of immunogenic chemotherapeutics.
Keywords: autophagy, ATP, calreticulin, dendritic cells, epothilone B, HMGB1
Introduction
The long-standing notion that apoptosis would invariably constitute an immunologically silent (and hence would fail to activate the immune system), if not a tolerogenic (and hence would actively suppress immune responses), cell death modality has recently been invalidated.1,2 Thus, at least in selected circumstances, cancer cells die while emitting a spatiotemporally defined combination of signals that results in the activation of tumor-specific cognate immune responses.3,4 The signals that have been associated with the productive perception of immunogenic cell death (ICD) by the immune system include, though presumably are not limited to, (1) the pre-apoptotic exposure of the endoplasmic reticulum (ER) chaperone calreticulin (CRT) and heat-shock proteins (HSPs), including HSP70 and HSP90, on the cell surface,5-8 (2) the autophagy-dependent active secretion of ATP9,10 and (3) the post-apoptotic release of the nuclear non-histone chromatin-binding protein high mobility group box 1 (HMGB1).11 By binding to CD91 on the surface of dendritic cells (DCs), CRT functions as an “eat-me” signal, thus promoting the engulfment of apoptotic corpses.5,12 The presence of HSPs on the surface or in the proximity of dying cancer cells results in the formation of tumor antigen-HSP complexes, which are processed by DCs for the cross-priming of T cells more efficiently than tumor antigens alone.13 Extracellular ATP stimulates the NLRP3 inflammasome, a platform for the activation of caspase-1,14 via purinergic P2RX7 receptors, hence promoting the proteolytic maturation and the release of pro-inflammatory cytokines such as interleukin (IL)-1β and IL-18.15 Finally, HMGB1 exerts immunogenic functions via a Toll-like receptor 4 (TLR4)- and MYD88-dependent signaling cascade that boosts the processing and cross-presentation of antigens from dying tumor cells.11 The molecular and cellular cascades that allow for the emission of immunogenic signals by dying cancer cells—on one hand—and for their perception by the immune system—on the other hand—have been intensively investigated. A detailed discussion of these mechanistic aspects however exceeds the scope of this Trial Watch and can be found elsewhere.3,16
To date, a rather restricted number of stimuli has been shown to induce bona fide ICD, hence converting dying cancer cells into a vaccine that elicits protective tumor-specific immune responses.3 These include some types of radiotherapy and photodynamic therapy,12,17-19 cardiac glycosides20,21 and a few chemotherapeutics: the DNA alkylating agent cyclophosphamide,22 the anthracyclines doxorubicin and mitoxantrone,5,23,24 the platinum derivative oxaliplatin,25 the microtubular poison patupilone (also known as epothilone B or EPO906)26 and the epidermal-growth factor receptor (EGFR)-targeting monoclonal antibody 7A7.27 The histone deacetylase inhibitor vorinostat as well as the n3-polyunsaturated fatty acid docosahexaenoic acid have been shown to promote the exposure of CRT on brain, pancreatic and bladder cancer cell lines.28,29 Along similar lines, bortezomib, an inhibitor of the proteasome nowadays approved by FDA for cancer therapy, reportedly stimulates the exposure of HSP90 on the surface of dying multiple myeloma cells,30 and the oncolytic coxsackievirus B3 has been suggested to kill non-small cell lung carcinoma (NSCLC) cells while allowing for the exposure of CRT on the cell surface, the secretion of ATP and the release of HMGB1.31 Finally, it has recently been demonstrated that neoplastic cells succumbing to shikonin, a phytochemical endowed with anti-inflammatory and antitumor properties, not only emits the three main signals associated with ICD abut also increase the immunogenic potential of DC-based vaccines.32,33 However, none of these observations has been confirmed with appropriate vaccination assays in vivo (see below),16 casting doubts on the hypothesis that vorinostat, docosahexaenoic acid, bortezomib, coxsackievirus B3 and shikonin would constitute bona fide ICD inducers. In 2012, we have demonstrated that hyperploidy, be it spontaneous or triggered by cytoskeletal poisons such as cytochalasin D or dihydrocytochalasin B, is associated with increased baseline levels of CRT exposure on the cell surface.26 Although this underpins an immunosurveillance mechanism that controls the ploidy of cancer cells in vivo, neither of these chemicals were shown to induce bona fide ICD, at least at the hyperploidizing concentrations employed in our study.26
Importantly, there appears to be no simple structure-function relationship between chemotherapeutics and their capacity to induce ICD, as chemically similar agents such as cisplatin and oxaliplatin do not behave similarly in this sense.25,34 This implies that the capacity of a given chemical to promote ICD cannot be predicted based on its structural analogy with a known ICD inducer, but rather must be assessed experimentally. We have recently developed fluorescent biosensors to monitor CRT exposure, ATP secretion and HMGB1 release in a high-throughput setting, allowing for the relatively straightforward identification of chemicals that induce the major hallmarks of ICD.20 Nevertheless, the true potential of a given stimulus to promote ICD must be confirmed in vivo, in assays that specifically evaluate the capacity of dying cancer cells to induce a protective immune response against live cells of the same type.3
Along the lines of our monthly Trial Watch series,35-45 here we will summarize the latest advances in the use of bona fide ICD inducers as anticancer agents, focusing on high-impact studies that have been published and clinical trials that have been launched during the last 13 months. Of note, the approval status of these agents has not varied since the publication of the latest Trial Watch dealing with this topic.44 Thus, (1) cyclophosphamide, doxorubicin and oxaliplatin are nowadays approved by FDA and other regulatory agencies for the treatment of distinct hematologic and solid malignancies, (2) mitoxantrone is predominantly employed for a cancer-unrelated indication, multiple sclerosis, even though mitoxantrone-containing chemotherapeutic regimens have also been endorsed by FDA for use in patients affected by acute myeloid leukemia, non-Hodgkin’s lymphoma, breast and prostate carcinoma and (3) patupilone remains an investigational agent (Table 1).
Table 1. Approved indications for immunogenic chemotherapy*,**.
Drug | Indications |
---|---|
Cyclophosphamide |
ALL, AML, breast cancer, CLL, CML, lupus nephritis, lymphoma, MM, mycosis fungoides, neuroblastoma, nephrotic syndrome, ovarian cancer, retinoblastoma. |
Doxorubicin |
ALL, AML, breast cancer, bronchogenic carcinoma, cervical carcinoma, gastric carcinoma, germ cell tumors, hepatocellular carcinoma, HNC, lymphoma, mesothelioma, MM, neuroblastoma, ovarian carcinoma, pancreatic carcinoma, prostate cancer, SCLC, soft tissue and bone sarcomas, thyroid carcinoma, transitional cell bladder carcinoma, uterine carcinoma, Wilms' tumor. |
Mitoxantrone |
Acute leukemia, breast cancer, NHL, multiple sclerosis, prostate cancer. |
Oxaliplatin |
Metastatic colorectal cancer. |
Patupilone | Investigational agent. |
Abbreviations: ALL, acute lymphocytic leukemia; AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; CML, chronic myeloid leukemia; MM, multiple myeloma; HNC, head and neck cancer; NHL, non-Hodgkin’s lymphoma; SCLC, small cell lung cancer
by FDA or European Medicines Agency (EMA) at the day of submission
updated from ref. 44
Of note, 7A7 is a murine IgG1 originally raised against murine EGFR, though it displays a consistent degree of cross-reactivity against human EGFR.46 Since multiple agents that specifically target human EGFR are currently available, including FDA-approved molecules (e.g., the tyrosine kinase inhibitor erlotinib, the monoclonal antibody cetuximab)47-49 as well as a wealth of investigational compounds (e.g., the monoclonal antibodies necitumumab and nimotuzumab),38,41 it seems very unlikely that the development of 7A7 will ever be pursued until a clinical stage. Unfortunately, for the reasons discussed above, no predictions can be put forward on the ability of other EGFR-targeting agents to stimulate bona fide ICD.50
Literature Update
Since the submission of our previous Trial Watch on this topic (December 2011),44 high-impact journals (latest impact factor > 15, according to Thomson Reuters–ISI Web of Knowledge) dealing with clinical cancer research (i.e., Science, Nature, Nature Medicine, The New England Journal of Medicine, Lancet, Lancet Oncology and The Journal of Clinical Oncology) have published the results of no less than 100 clinical trials involving the use of ICD inducers in cancer patients. In the vast majority of cases, however, cyclophosphamide, doxorubicin, oxaliplatin and mitoxantrone were not evaluated for novel therapeutic indications but administered to the control arm of patient cohorts as part of gold standard therapeutic regimens. Thus, during the last 13 months, a relatively limited number of high-impact publications reported the results of clinical studies aimed at investigating the use of FDA-approved ICD inducers (mitoxantrone excluded) as off-label anticancer medications. Alongside, one single article dealt with the potential antineoplastic activity of patupilone (source www.ncbi.nlm.nih.gov/sites/entrez/).
Combinatorial chemotherapeutic regimens involving metronomic cyclophosphamide have been tested in two Phase I clinical trials enrolling cancer patients of different ages.51,52 Thus, the combination of metronomic cyclophosphamide with the poly(ADP-ribose) polymerase (PARP) inhibitor velaparib turned out to be well tolerated by adults affected by refractory solid tumors and to exert promising antineoplastic effects in a subset of patients bearing BRCA1/2 mutations.51 Along similar lines, low-dose cyclophosphamide combined with the vascular endothelial growth factor (VEGF)-specific monoclonal antibody bevacizumab53 and the FDA-approved multi-kinase inhibitor sorafenib54 induced no significant toxicity (and exerted antineoplastic activity, at least to some extent) in children and young adults affected by refractory/recurrent solid tumors.52 In addition, it has recently been shown that metronomic cyclophosphamide efficiently reduce the number of immunosuppressive FOXP3+ regulatory T cells in the circulation of advanced renal cell carcinoma patients, allowing for the development of antitumor responses upon the administration of a multipeptide vaccine formulation (IMA901).55
As an off-label anticancer agent, doxorubicin has been recently tested (1) in combination with the cyclin-dependent kinase (CDK) inhibitor flavopiridol in patients affected by advanced sarcomas,56 (2) as a non-targeted liposomal formulation and combined with bevacizumab plus temsirolimus (an inhibitor of the mammalian target of rapamycin, mTOR, approved by FDA for use in renal cell carcinoma patients)57 in individuals bearing advanced malignancies58 and (3) as the chemotherapeutic cargo of EGFR-targeted immunoliposomes in patients affected by advanced solid tumors.59 All these doxorubicin-based chemotherapeutic regimens were well tolerated and exerted (at least partial) antineoplastic effects, supporting the initiation of Phase II studies. Furthermore, the combination of doxorubicin, cisplatin and etoposide (a topoisomerase II inhibitor) has been shown to ameliorate the response rate and progression-free survival (but not the overall survival) of advanced adrenocortical carcinoma patients receiving mitotane (a derivative of the organochloride insecticide DDT currently approved by FDA for use in these patients) more consistently than streptozocin (a natural alkylating agent nowadays employed against pancreatic cancer).60
The most remarkable progress in the clinical development of oxaliplatin achieved since the submission of our latest Trial Watch (December 2011)44 surely involve pancreatic cancer.61-63 The FOLFIRINOX regimen (folinic acid + 5-fluorouracil + irinotecan + oxaliplatin) has indeed been shown not only to increase the response rate but also to extend the overall survival (by more than 4 months) of individuals affected by metastatic pancreatic cancer, as compared with the nucleoside analog gemcitabine.61-64 Although this chemotherapeutic regimen was also associated with an increased rate of adverse events,62 it has generated a considerable clinical interest, as demonstrated by the consistent number (namely, 21) of clinical trials initiated during the last 13 months to test the antineoplastic profile of oxaliplatin-based chemotherapy in cohorts of pancreatic cancer patients (see below). Very encouraging results have been obtained in two Phase III clinical trials testing (1) the XELOX (capecitabine + oxaliplatin) regimen as an adjuvant intervention for patients subjected to curative D2 gastroctemy65 and (2) the GEMOX (gemcitabine + oxaliplatin) regimen, as a standalone intervention or combined with erlotinib, in patients affected by biliary-tract cancer.66 In line with this notion, no less than 21 clinical trials have recently been initiated to test the antineoplastic potential of oxaliplatin-based chemotherapeutic regimens in cohorts of patients affected by gastric, gastresophageal, gastrointestinal and biliary duct neoplasms.
During the last 13 months, high-impact journals have published one single article dealing with the clinical anticancer potential of patupilone. In this work, Colombo and colleagues report the results of a randomized, open-label, Phase III study comparing patupilone with pegylated liposomal doxorubicin (PLD) in platinum-refractory patients affected by recurrent ovarian carcinoma, primary fallopian tube cancer or primary peritoneal cancer.67 A total of 829 individuals were randomly assigned to receive either 10 mg/m2 patupilone (i.v., every 3 weeks) or 50 mg/m2 PLD (i.v., every 4 weeks) and monitored for clinical progression and the emergence of adverse effects. Of note, although a higher rate of partial responses was recorded in the experimental arm, patupilone did not significantly ameliorate overall survival (the primary end-point of the study) as compared with PLD.67
Focusing on recent, high-impact translational research focusing on ICD-inducing chemotherapeutics, we have found of particular interest the works by Senovilla et al., Dewan et al., Qayum et al. and Zhang et al., demonstrating (1) that patupilone is capable of triggering ICD;26 (2) that low-dose cyclophosphamide and irradiation (both constituting bona fide ICD inducers) synergize with the FDA-approved TLR7 agonist imiquimod in inhibiting tumor growth in an ectopic murine model of breast carcinoma, an effect that is associated with abundant tumor infiltration by CD11c+, CD4+ and CD8+ cells; (3) that a chemical inhibitor of class I phosphoinositide-3-kinases (i.e., GDC-0941) normalizes the tumor vasculature in vivo, hence increasing perfusion, restoring normoxia and allowing for the delivery of increased amounts of doxorubicin;68 and (4) that doxorubicin—contrarily to long-established convictions—does not exert cardiotoxic effects as it directly stimulates the mitochondrial generation of reactive oxygen species (ROS), but rather by specifically targeting topoisomerase IIβ (at least in mice).69
Update on Clinical Trials
When this Trial Watch was being redacted (December 2012), official sources listed no less than 305 clinical trials launched after December 1, 2011, that would investigate the safety and therapeutic profile of bona fide ICD inducers in cancer patients (source www.clinicaltrials.gov). Of these, 83 studies are based on (or at least include among other therapeutic interventions) cyclophosphamide, 63 doxorubicin, 80 oxaliplatin and 9 mitoxantrone. Of note, no clinical trials involving patupilone have been registered at www.clinicaltrials.gov during the last 13 months. An unrestricted search for the term “patupilone” returns 59 entries; yet only 29 of these actually involved patupilone. Virtually all clinical studies testing patupilone in cancer patients have nowadays been completed, suspended, withdrawn or terminated, as they were near-to-invariably initiated before 2008.
Among 83 recently initiated clinical trials investigating the safety and therapeutic profile of cyclophosphamide in cancer patients, 61 involve only indications for which this drug has already been approved by FDA (Table 1), including breast cancer (23 trials) and a wide panel of hematological malignancies (38 trials) (source www.clinicaltrials.gov). In addition, cyclophosphamide is currently being tested in patients affected by: (1) melanoma (NCT01495572; NCT01585415; NCT01659151; NCT01701674; NCT01740401; NCT01740557), in combination with adoptive cell transfer, or the FDA-approved anti-cytotoxic T-lymphocyte antigen 4 (CTLA4) monoclonal antibody ipilimumab;70,71 (2) pediatric solid (including brain) tumors (NCT01535183; NCT01661400; NCT01670175), as a standalone intervention or combined with either conventional chemotherapeutic regimens or rapamycin (another immunosuppressive inhibitor of mTOR)72 plus topotecan (a topoisomerase inhibitor); (3) hematological malignancies (NCT01509300; NCT01685411; NCT01690520), invariably as part of a lymphodepleting/lymphoablative conditioning regimen that precedes hematopoietic stem cell transplantation; (4) sarcoma (NCT01492673; NCT01716689), as a single agent or in association with topotecan and bevacizumab; (5) head and neck cancer (NCT01581970), in combination with cetuximab;49 (6) mesothelioma and pancreatic cancer (NCT01583686), as a part of a conditioning regimen followed by the administration of peripheral blood lymphocytes engineered to express an anti-mesothelin chimeric antigen receptor (CAR);73-75 (7) pancreatic cancer (NCT01595321), as an immunostimulatory intervention to maximize the efficacy of a tumor-cell vaccine; (8) prostate cancer (NCT01696877), combined with androgen ablation and a granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transfected tumor-cell vaccine; (9) rectal carcinoma (NCT01507103), in association with chemotherapy and a liposomal vaccine that targets the exposed core domain of mucin 1 (L-BLP25); (10) reproductive tract neoplasms (NCT01606241), as an immunostimulatory intervention in support of a folate receptor α (FRα)-targeting vaccine; and (11) not better defined advanced solid tumors (NCT01598129; NCT01697527), either as part of a lymphodepleting regimen preceding the infusion of genetically engineered lymphocytes or combined with an oncolytic adenovirus (CGTG-02) (Table 2).
Table 2. Clinical trials recently launched to evaluate the antineoplastic profile of cyclophosphamide as an off-label medication*.
Indications | Status | Phase | Notes | Ref. |
---|---|---|---|---|
CML MDS Solid tumors |
Not yet recruiting |
n.a. |
Combined with busulfan and followed by allogeneic HSCT |
NCT01685411 |
II |
Combined with fludarabine, TBI and followed by UCBT |
NCT01690520 |
||
Recruiting |
I/II |
Combined with fludarabine, TBI and followed by HSCT |
NCT01509300 |
|
Ewing’s sarcoma Neuroblastoma |
Recruiting |
II |
Combined with topotecan and bevacizumab |
NCT01492673 |
HNC |
Recruiting |
II |
Combined with cetuximab |
NCT01581970 |
Melanoma |
Not yet recruiting |
I/II |
Combined with fludarabine, ACT and rIL-2 |
NCT01740557 |
Recruiting |
n.a. |
Combined with fludarabine, ACT, rIL-2 and ipilimumab |
NCT01701674 |
|
I |
Combined with fludarabine, ACT, rIL-2 and vemurafenib |
NCT01585415 |
||
II |
Combined with ACT and vemurafenib |
NCT01659151 |
||
Combined with fludarabine, ACT and rIL-2 |
NCT01495572 |
|||
Combined with ipilimumab |
NCT01740401 |
|||
Mesothelioma Pancreatic cancer |
Recruiting |
II |
Combined with fludarabine and ACT with anti-mesothelin PBLs |
NCT01583686 |
Pancreatic cancer |
Recruiting |
n.a. |
FOLFIRINOX regimen combined with SBRT and tumor-cell vaccine |
NCT01595321 |
Pediatric solid tumors |
Not yet recruiting |
n.a. |
As single agent |
NCT01661400 |
II |
Combined with carboplatin, etoposide, irinotecan and vincristine |
NCT01535183 |
||
Recruiting |
I |
Combined with rapamycin and topotecan |
NCT01670175 |
|
Prostate cancer |
Recruiting |
I/II |
Combined with androgen ablation and a GM-CSF-expressing tumor-cell vaccine |
NCT01696877 |
Rectal carcinoma |
Recruiting |
II |
Combined with chemoradiotherapy and a MUC1-targeting vaccine |
NCT01507103 |
Reproductive tract cancers |
Recruiting |
I |
Combined with a FRα-targeting vaccine |
NCT01606241 |
Sarcoma |
Recruiting |
II |
As single agent |
NCT01716689 |
Solid tumors | Recruiting | I |
Combined with an oncolytic virus |
NCT01598129 |
II | Combined with fludarabine, irradiation, ACT and rIL-2 | NCT01697527 |
Abbreviations: ACT, adoptive cell transfer; CML, chronic myelogenous leukemia; FOLFIRINOX, folinic acid, 5-fluorouracil, irinotecan, oxaliplatin; FRα, folate receptor α; GM-CSF, granulocyte-macrophage colony-stimulating factor; HNC, head and neck cancer; HSCT, hematopoietic stem cell transplantation; MDS, myelodysplastic syndrome; MUC1, mucin 1; n.a., not available, rIL-2, recombinant interleukin-2; PBL, peripheral blood lymphocytes; SBRT, stereotactic body radiation; TBI, total body irradiation; UCBT, umbilical cord blood transplantation
between December 1, 2011, and the day of submission
Forty-nine out of 63 clinical studies recently launched to evaluate the safety profile and antineoplastic activity of doxorubicin in cancer patients involve only indications for which this compound is nowadays approved by FDA (Table 1), including breast carcinoma (15 trials), hepatocellular carcinoma (6 trials), ovarian carcinoma (3 trials), soft tissue sarcoma (2 trials), pancreatic carcinoma (1 trial) and a wide array of hematological tumors (22 trials) (source www.clinicaltrials.gov). Furthermore, doxorubicin is being assessed as an antineoplastic agent in cohorts of patients bearing: (1) neoplasms of the reproductive tract (NCT01489371; NCT01659554; NCT01666444; NCT01673477), most often as PLD combined with cisplatin or with immunostimulatory interventions encompassing TLR8 agonists and an IL-12-encoding plasmid; (2) sarcomas or leiomyosarcomas (NCT01490060; NCT01533207; NCT01746238), in combination with conventional chemotherapeutic regimens, bevacizumab plus radiotherapy or gemcitabine plus granulocyte colony-stimulating factor (G-CSF); (3) chronic myelogenous leukemia (NCT01670084), as a part of a combinatorial therapy involving—among other drugs—cytarabine (a nucleoside analog), corticosteroids and methotrexate (an antifolate); (4) bladder and urothelial cancer (NCT01639521), combined with cisplatin, vinblastine (a microtubular poison) and methotrexate; (5) metastatic colorectal carcinoma (NCT01703910), as a standalone intervention; (6) hepatic metastases from digestive endocrine neoplasms (NCT01678664), in the context of chemoembolization and followed by the administration of everolimus (an immunosuppressive derivative of rapamycin); and (7) not better defined advanced or metastatic solid tumors (NCT01593228; NCT01640847; NCT01673438), in the form of both free and tumor-targeted drug, combined with carboplatin (a platinum compound related, but not equivalent to, cisplatin and oxaliplatin)25,76,77 plus iniparib (a compound originally developed as—but later disconfirm from being—an irreversible inhibitor of PARP1),78,79 or coupled to high intensity focused ultrasound (HIFU) (Table 3).
Table 3. Clinical trials recently launched to evaluate the antineoplastic profile of doxorubicin as an off-label medication*.
Indications | Status | Phase | Notes | Ref. |
---|---|---|---|---|
Advanced or metastatic solid tumors |
Not yet recruiting |
II |
Combined with HIFU |
NCT01640847 |
Recruiting |
I/II |
Combined with aldoxorubicin |
NCT01673438 |
|
III |
As liposomal injection combined with carboplatin and iniparib |
NCT01593228 |
||
CML |
Not yet recruiting |
II |
Combined with various drugs including araC, folonic acid and metotrexate |
NCT01670084 |
Bladder cancer Urothelial cancer |
Not yet recruiting |
II |
MVAC regimen combined with G-CSF |
NCT01639521 |
Colorectal carcinoma |
Not yet recruiting |
II |
As single agent |
NCT01703910 |
Hepatic metastases of neuroendocrine tumors |
Not yet recruiting |
II |
Combined with chemoembolization and everolimus |
NCT01678664 |
Leiomyosarcoma |
Recruiting |
III |
TAC regimen combined with gemcitabine and G-CSF |
NCT01533207 |
Sarcoma |
Not yet recruiting |
n.a. |
Combined with various drugs including dexamethasone and vincristine |
NCT01490060 |
Recruiting |
I |
Combined with bevacizumab and radiotherapy |
NCT01746238 |
|
Reproductive tract cancers | Not yet recruiting |
I |
As PLD and combined with EGEN-001 |
NCT01673477 |
Recruiting | I |
As PLD and combined with EGEN-001 |
NCT01489371 |
|
II | Combined with cisplatin |
NCT01659554 |
||
As PLD and combined with TLR8 agonist | NCT01666444 |
Abbreviations: araC, cytarabine; CML, chronic myelogenous leukemia; G-CSF, granulocyte colony-stimulating factor; HIFU, high intensity focused ultrasound; MVAC, cisplatin, vinblastine, doxorubicin, methotrexate; n.a., not available; PLD, pegylated liposomal doxorubicin; TAC, docetaxel, doxorubicin, cyclophosphamide; TLR8, Toll-like receptor 8
between December 1, 2011, and the day of submission
Among 80 clinical trials recently launched to estimate the safety and efficacy of oxaliplatin in cancer patients, 30 are enrolling/will enroll only individuals affected by colorectal carcinoma, the sole indication for which this platinum-derivative is currently approved by FDA and other international regulatory agencies (Table 1) (source www.clinicaltrials.gov). In addition, oxaliplatin is currently being tested for its antineoplastic properties in cohorts of patients bearing: (1) pancreatic cancer (NCT01485744; NCT01521702; NCT01524575; NCT01526135; NCT01558869; NCT01560949; NCT01581307; NCT01586611; NCT01591733; NCT01595321; NCT01632306; NCT01643499; NCT01652976; NCT01658943; NCT01660711; NCT01666730; NCT01671202; NCT01683422; NCT01688336; NCT01744353; NCT01571024), near-to-invariably as part of conventional GEMOX, FOLFOX (folinic acid + 5-fluorouracil + oxaliplatin) or FOLFIRINOX regimens; (2) gastric, gastresophageal or gastrointestinal tumors (NCT01515748; NCT01516944; NCT01534546; NCT01552980; NCT01558011; NCT01583361; NCT01590719; NCT01618474; NCT01630083; NCT01662869; NCT01665274; NCT01671449; NCT01711242; NCT01643499; NCT01608646; NCT01693445; NCT01498289; NCT01523015; NCT01732380; NCT01747551; NCT01719926), most often as part of established FOLFOX, DOS (docetaxel + oxaliplatin), SOX (S-1 + oxaliplatin) or XELOX regimens; (3) B-cell lymphoma (NCT01562990, NCT01670370), in the context of the GEMOX treatment alone or further combined with the FDA-approved anti-CD20 monoclonal antibody rituximab;80,81 (4) breast carcinoma (NCT01528826; NCT01658033), combined with vinorelbine (a vinca alkaloid) or in the context of the FOLFOX regimen plus bevacizumab; (5) cholangiocarcinoma (NCT01525069; NCT01572324), combined with gemcitabine, dexamethasone and floxuridine (a nucleoside analog) or as part of the FOLFOX treatment plus capecitabine; (6) biliary tract neoplasms (NCT01494363; NCT01643499), as part of the FOLFIRINOX regimen; (7) peritoneal carcinomatosis (NCT01575730), as a standalone chemotherapeutic intervention, given as normothermic vs. hyperthermic intraoperative chemoperfusion; and (8) not better specified solid tumors (NCT01611857), in the context of the FOLFOX treatment further combined with tivantinib (a hitherto experimental inhibitor of the oncogenic kinase MET)82 (Table 4).
Table 4. Clinical trials recently launched to evaluate the antineoplastic profile of oxaliplatin as an off-label medication*.
Indications | Status | Phase | Notes | Ref. |
---|---|---|---|---|
BCL |
Active, not recruiting |
I/II |
GEMOX regimen plus rituximab |
NCT01562990 |
Recruiting |
III |
GEMOX regimen |
NCT01670370 |
|
Biliary tract cancer |
Recruiting |
II |
FOLFIRINOX regimen |
NCT01494363 |
Biliary tract cancer Gastrointestinal cancer Pancreatic cancer |
Recruiting |
I |
FOLFIRINOX regimen |
NCT01643499 |
Breast carcinoma |
Recruiting |
II |
FOLFOX regimen plus bevacizumab |
NCT01658033 |
NVBOX regimen |
NCT01528826 |
|||
Cholangiocarcinoma |
Recruiting |
n.a. |
GEMOX regimen plus dexamethasone plus floxuridine |
NCT01525069 |
II |
FOLFOX regimen plus capecitabine |
NCT01572324 |
||
Gastric cancer |
Not yet recruiting |
II |
XELOX regimen |
NCT01665274 |
XELOX regimen followed by docetaxel |
NCT01558011 |
|||
III |
SOX or XELOX regimen |
NCT01534546 |
||
SOX regimen |
NCT01671449 |
|||
Recruiting |
II |
Adjuvant XELOX regimen |
NCT01618474 |
|
EOX regimen plus immunotherapy |
NCT01630083 |
|||
FOLFOX regimen plus onartuzumab |
NCT01590719 |
|||
SOX regimen |
NCT01552980 |
|||
II/III |
Adjuvant or perioperative SOX or XELOX regimen |
NCT01516944 |
||
III |
Adjuvant or neoadjuvant SOX regimen |
NCT01583361 |
||
DOS regimen |
NCT01515748 |
|||
FOLFOX regimen plus onartuzumab |
NCT01662869 |
|||
XELOX regimen plus radiotherapy |
NCT01711242 |
|||
Gastresophageal cancers |
Not yet recruiting |
II |
FOLFOX regimen plus anti-VEGF therapy |
NCT01747551 |
Recruiting |
I |
XELOX regimen |
NCT01719926 |
|
II |
Combined with radiotherapy and raltiterxed |
NCT01732380 |
||
FOLFOX regimen |
NCT01498289 |
|||
II/III |
Combined with docetaxel, 5-FU and radiotherapy |
NCT01523015 |
||
Gastrointestinal tumors |
Recruiting |
I |
OIS regimen |
NCT01693445 |
II |
SOX regimen |
NCT01608646 |
||
Pancreatic cancer |
Not yet recruiting |
I |
FOLFOX regimen plus PI3K inhibitor |
NCT01571024 |
I/II |
FOLFOX regimen plus LY2090314 |
NCT01632306 |
||
NCT01671202 | ||||
II |
FOLFOX regimen plus metformin |
NCT01666730 |
||
III |
FOLFOX regimen |
NCT01586611 |
||
Recruiting |
n.a. |
FOLFIRINOX regimen combined with SBRT and tumor-cell vaccine |
NCT01595321 |
|
I |
FOLFIRINOX plus LDE225 |
NCT01485744 |
||
FOLFOX-A regimen |
NCT01744353 |
|||
II |
GEMOX regimen |
NCT01524575 |
||
XELOX regimen plus irinotecan |
NCT01558869 |
|||
Neoadjuvant FOLFIRINOX regimen followed by gemcitabine-based therapy |
NCT01560949 |
|||
FOLFOX regimen plus theraspheres |
NCT01581307 |
|||
FOLFIRINOX regimen followed by capecitabine and radiotherapy |
NCT01591733 |
|||
FOLFOX regimen + dasatinib |
NCT01652976 |
|||
FOLFOX regimen |
NCT01658943 |
|||
FOLFIRINOX regimen |
NCT01660711 |
|||
XELOX regimen plus gemcitabine, erlotinib and proton radiation |
NCT01683422 |
|||
III |
Neoadjuvant GEMOX regimen |
NCT01521702 |
||
FOLFIRINOX regimen |
NCT01526135 |
|||
NCT01688336 | ||||
Peritoneal carcinomatosis |
Recruiting |
II |
As normothermic or hypothermic intraperitoneal chemotherapy |
NCT01575730 |
Solid tumors | Recruiting | I/II | FOLFOX regimen plus tivantinib | NCT01611857 |
Abbreviations: 5-FU, 5-fluorouracil; BCL, B-cell lymphoma; DOS, docetaxel + oxaliplatin; EOX, epirubicin + oxaliplatin + capecitabine; FOLFIRINOX, folinic acid + 5-FU + irinotecan + oxaliplatin; FOLFOX, folinic acid + 5-FU + oxaliplatin; GEMOX, gemcitabine + oxaliplatin; n.a., not available; NVBOX, vinorelbine + oxaliplatin; OIS, oxaliplatin + irinotecan + S-1; PI3K, phosphoinositide-3-kinase; SBRT, stereotactic body radiation therapy; SOX, S-1 + oxaliplatin; VEGF, vascular endothelial growth factor; XELOX, capecitabine + oxaliplatin
between December 1, 2011, and the day of submission
Of 9 clinical studies initiated after 2011, December 1 to assess the safety and efficacy of mitoxantrone in cancer patients, 6 involve only indications for which this anthracycline is currently approved by FDA (Table 1), including various types of leukemia (4 trials) and prostate cancer (2 trials) (source www.clinicaltrials.gov). Moreover, mitoxantrone is nowadays being investigated in patients affected by (1) myelodysplastic syndromes (NCT01701375; NCT01729845), combined with cytarabine and either decitabine (a hypomethylating agent) plus etoposide or an experimental inhibitor of CDK4 and CDK6 (PD 0332991);83 and (2) relapsed or refractory mantle cell lymphoma (NCT01578343), as a part of a combinatorial regimen including fludarabine (a nucleoside analog), vorinostat (an inhibitor of histone deacetylases) and corticosteroids (dexamethasone) (Table 5).
Table 5. Clinical trials recently launched to evaluate the antineoplastic profile of mitoxantrone as an off-label medication*.
Indications | Status | Phase | Notes | Ref. |
---|---|---|---|---|
MDS |
Not yet recruiting |
I/II |
Combined with araC, decitabine and etoposide |
NCT01729845 |
Recruiting |
I |
Combined with araC and PD 0332991 |
NCT01701375 |
|
MCL | Not yet recruiting | II | Combined with dexamethasone, fludarabine and vorinostat | NCT01578343 |
Abbreviations: araC, cytarabine; MCL, mantle cell lymphoma; MDS, myelodysplastic syndrome
between December 1, 2011, and the day of submission
Concluding Remarks
The concept of immunogenic cell death has originally been proposed in 2005 by Casares et al., who were the first (1) to demonstrate that malignant cells exposed to anthracyclines are capable of vaccinating mice against a subsequent challenge with tumor cells of the same type, and (2) to provide mechanistic insights into this phenomenon, i.e., to show that it depends on the activity of caspases.23 Since then, the molecular machineries whereby dying cancer cells emit a spatiotemporally defined combination of signals that—at least under selected circumstances—can be translated by the immune system into an antitumor response have been intensively investigated.5,6,10,12,20 Alongside, the list of bona fide ICD inducers has been significantly expanded, though at a relatively slow pace. One of the major obstacles to the identification of novel chemicals that trigger bona fide ICD is that this property cannot be easily predicted starting from structural features, but rather must be tested experimentally. As a standalone example, cisplatin and oxaliplatin, two structurally related platinum derivatives that ignite largely redundant (though not precisely overlapping) signaling pathways and activate highly similar (though not identical) resistance mechanisms,76 differ in their capacity to induce ICD.25,34 In addition, the immunogenic potential of some chemotherapeutics is largely influenced by dose and administration schedule. Thus, while high-dose cyclophosphamide is well known for its potent immunosuppressive activity, and has been used for years in this sense (for instance as part of lymphodepleting/lymphoablative regimens to condition transplantation recipients), the same drug administered metronomically exerts a wide array of immunostimulatory functions.22,44,84,85 Thus, even though high-throughput screening-compatible systems allowing for the evaluation of the major hallmarks of ICD (i.e., CRT exposure, ATP secretion, HMGB1 release) are nowadays available,20 the gold standard method to assess the ability of a given stimulus to trigger ICD is constituted by vaccination assays in immunocompetent, syngenic animals.3,16 Besides being cost-ineffective and relatively time-consuming, this approach turns out to be particularly disadvantageous when murine cells are resistant to experimental manipulations that “normally” affect their human counterparts, e.g., cardiac glycosides.20,21,39 It is tempting to speculate, yet remains to be formally demonstrated, that the use of mice genetically engineered to bear a virtually complete human immune system (so-called humanized mice)86 may circumvent—at least in part—this issue.
One of the most recently identified ICD inducers is patupilone,26 a macrolide of the epothilone family originally described in 1995 for its ability to stabilize microtubules similar to taxol.87-89 The synthesis and preclinical characterization of epothilones generated an intense wave of enthusiasm,90,91 rapidly translating into dozens of clinical studies.92-95 In 2007, the U.S. FDA approved ixabepilone (a derivative of patupilone) for use as a standalone intervention in anthracycline-, taxane- and capecitabine-resistant breast carcinoma patients, or combined with capecitabine in patients bearing anthracycline- and taxane-resistant locally advanced or metastatic breast carcinoma.96,97 This perhaps explains why the clinical interest in patupilone appears to have nowadays completely declined (see above). Unfortunately, to the best of our knowledge, whether ixabepilone also constitutes a bona fide ICD inducer has not yet been determined, nor it has been investigated to which extent the immune system contributes to the clinical efficacy of this epothilone. Future studies will have to address these two therapeutically relevant questions.
In addition, it will be crucial to identify (1) therapeutic interventions that convert non-immunogenic apoptosis into ICD, as well as (2) biomarkers that can be used to predict the propensity of the patient’s immune system to productively detect ICD and translate it into an anticancer immune response. Some progress has already been made in this sense. On one hand, it has been demonstrated that cisplatin-induced cell death, which per se is associated with the secretion of ATP and the release of HGMB1 but not with the exposure of CRT on the cell surface, can be rendered immunogenic by the adsorption of recombinant CRT on dying cells as well as by the co-administration of the ER stressors such as thapsigargin and tunicamycin.5,34 Along similar lines, autophagy-deficient cancer cells succumbing to anthracyclines (i.e., bona fide ICD inducers) are intrinsically unable to promote ICD as they do not release ATP.9,10 At least speculatively, this represents a clinically unfavorable situation, which nevertheless can be reverted by the local administration of extracellular ATPase inhibitors such as suramine.10 On the other hand, breast carcinoma patients bearing loss-of-function polymorphisms in TLR4 and P2RX7 have been shown to relapse more quickly upon anthracycline-based chemotherapy and radiotherapy than control patients bearing wild-type polymorphisms.11,15
As it stands, the capacity of a given chemotherapeutic agent to promote ICD is only the first conditio sine qua non for the elicitation of therapeutic anticancer immune response. In addition, (1) the molecular machinery underpinning the emission of immunogenic signals by dying cancer cells must fully functional and (2) the patient’s immune system must be able to detect these signals, decode them and eventually orchestrate a cytotoxic T-cell response. Investigating in detail these three “modules” (i.e., the ICD inducer, the cancer cell and the patient’s immune system) and how they mutually interact will undoubtedly drive the discovery of novel ICD inducers as well as of clinically meaningful strategies to endow originally non-immunogenic instances of cell death with potent immunogenic properties.
Acknowledgments
Authors are supported by the European Commission (ArtForce); Agence National de la Recherche (ANR); Ligue contre le Cancer (Equipe Labelisée); Fondation pour la Recherche Médicale (FRM); Institut National du Cancer (INCa); LabEx Immuno-Oncologie; Fondation de France; Fondation Bettencourt-Schueller; AXA Chair for Longevity Research; Cancéropôle Ile-de-France and Paris Alliance of Cancer Research Institutes (PACRI).
Glossary
Abbreviations:
- CAR
chimeric antigen receptor
- CDK
cyclin-dependent kinase
- CRT
calreticulin
- CTLA4
cytotoxic T-lymphocyte antigen 4
- DC
dendritic cell
- DOS
docetaxel + oxaliplatin
- EGFR
epidermal growth factor receptor
- ER
endoplasmic reticulum
- FOLFIRINOX
folinic acid + 5-fluorouracil + irinotecan + oxaliplatin
- FOLFOX
folinic acid + 5-fluorouracil + oxaliplatin
- FRα
folate receptor α
- G-CSF
granulocyte colony-stimulating factor
- GEMOX
gemcitabine + oxaliplatin
- GM-CSF
granulocyte-macrophage colony-stimulating factor
- HIFU
high intensity focused ultrasound
- HMGB1
high mobility group box 1
- HSP, heat-shock protein
ICD, immunogenic cell death
- IL
interleukin
- mTOR
mammalian target of rapamycin
- NSCLC
non-small cell lung carcinoma
- PARP
poly(ADP-ribose) polymerase 1
- PLD
pegylated liposomal doxorubicin
- ROS
reactive oxygen species
- SOX
S-1 + oxaliplatin
- TLR
Toll-like receptor
- VEGF
vascular endothelial growth factor
- XELOX
capecitabine + oxaliplatin
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Footnotes
† These authors share senior co-authorship.
Previously published online: www.landesbioscience.com/journals/oncoimmunology/article/23510
References
- 1.Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012;19:107–20. doi: 10.1038/cdd.2011.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Galluzzi L, Maiuri MC, Vitale I, Zischka H, Castedo M, Zitvogel L, et al. Cell death modalities: classification and pathophysiological implications. Cell Death Differ. 2007;14:1237–43. doi: 10.1038/sj.cdd.4402148. [DOI] [PubMed] [Google Scholar]
- 3.Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2012 doi: 10.1146/annurev-immunol-032712-100008. In press. [DOI] [PubMed] [Google Scholar]
- 4.Zitvogel L, Kepp O, Kroemer G. Decoding cell death signals in inflammation and immunity. Cell. 2010;140:798–804. doi: 10.1016/j.cell.2010.02.015. [DOI] [PubMed] [Google Scholar]
- 5.Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13:54–61. doi: 10.1038/nm1523. [DOI] [PubMed] [Google Scholar]
- 6.Panaretakis T, Kepp O, Brockmeier U, Tesniere A, Bjorklund AC, Chapman DC, et al. Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell death. EMBO J. 2009;28:578–90. doi: 10.1038/emboj.2009.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Melcher A, Todryk S, Hardwick N, Ford M, Jacobson M, Vile RG. Tumor immunogenicity is determined by the mechanism of cell death via induction of heat shock protein expression. Nat Med. 1998;4:581–7. doi: 10.1038/nm0598-581. [DOI] [PubMed] [Google Scholar]
- 8.Srivastava P. Roles of heat-shock proteins in innate and adaptive immunity. Nat Rev Immunol. 2002;2:185–94. doi: 10.1038/nri749. [DOI] [PubMed] [Google Scholar]
- 9.Martins I, Tesniere A, Kepp O, Michaud M, Schlemmer F, Senovilla L, et al. Chemotherapy induces ATP release from tumor cells. Cell Cycle. 2009;8:3723–8. doi: 10.4161/cc.8.22.10026. [DOI] [PubMed] [Google Scholar]
- 10.Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 2011;334:1573–7. doi: 10.1126/science.1208347. [DOI] [PubMed] [Google Scholar]
- 11.Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–9. doi: 10.1038/nm1622. [DOI] [PubMed] [Google Scholar]
- 12.Garg AD, Krysko DV, Verfaillie T, Kaczmarek A, Ferreira GB, Marysael T, et al. A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. EMBO J. 2012;31:1062–79. doi: 10.1038/emboj.2011.497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Castellino F, Boucher PE, Eichelberg K, Mayhew M, Rothman JE, Houghton AN, et al. Receptor-mediated uptake of antigen/heat shock protein complexes results in major histocompatibility complex class I antigen presentation via two distinct processing pathways. J Exp Med. 2000;191:1957–64. doi: 10.1084/jem.191.11.1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zitvogel L, Kepp O, Galluzzi L, Kroemer G. Inflammasomes in carcinogenesis and anticancer immune responses. Nat Immunol. 2012;13:343–51. doi: 10.1038/ni.2224. [DOI] [PubMed] [Google Scholar]
- 15.Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med. 2009;15:1170–8. doi: 10.1038/nm.2028. [DOI] [PubMed] [Google Scholar]
- 16.Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 2012;12:860–75. doi: 10.1038/nrc3380. [DOI] [PubMed] [Google Scholar]
- 17.Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, et al. Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood. 2009;114:589–95. doi: 10.1182/blood-2009-02-206870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Korbelik M, Zhang W, Merchant S. Involvement of damage-associated molecular patterns in tumor response to photodynamic therapy: surface expression of calreticulin and high-mobility group box-1 release. Cancer Immunol Immunother. 2011;60:1431–7. doi: 10.1007/s00262-011-1047-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sanovic R, Verwanger T, Hartl A, Krammer B. Low dose hypericin-PDT induces complete tumor regression in BALB/c mice bearing CT26 colon carcinoma. Photodiagnosis Photodyn Ther. 2011;8:291–6. doi: 10.1016/j.pdpdt.2011.04.003. [DOI] [PubMed] [Google Scholar]
- 20.Menger L, Vacchelli E, Adjemian S, Martins I, Ma Y, Shen S, et al. Cardiac glycosides exert anticancer effects by inducing immunogenic cell death. Sci Transl Med. 2012;4:43ra99. doi: 10.1126/scitranslmed.3003807. [DOI] [PubMed] [Google Scholar]
- 21.Kepp O, Menger L, Vacchelli E, Adjemian S, Martins I, Ma Y, et al. Anticancer activity of cardiac glycosides: At the frontier between cell-autonomous and immunological effects. Oncoimmunology. 2012;1:1640–2. doi: 10.4161/onci.21684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Schiavoni G, Sistigu A, Valentini M, Mattei F, Sestili P, Spadaro F, et al. Cyclophosphamide synergizes with type I interferons through systemic dendritic cell reactivation and induction of immunogenic tumor apoptosis. Cancer Res. 2011;71:768–78. doi: 10.1158/0008-5472.CAN-10-2788. [DOI] [PubMed] [Google Scholar]
- 23.Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med. 2005;202:1691–701. doi: 10.1084/jem.20050915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Fucikova J, Kralikova P, Fialova A, Brtnicky T, Rob L, Bartunkova J, et al. Human tumor cells killed by anthracyclines induce a tumor-specific immune response. Cancer Res. 2011;71:4821–33. doi: 10.1158/0008-5472.CAN-11-0950. [DOI] [PubMed] [Google Scholar]
- 25.Tesniere A, Schlemmer F, Boige V, Kepp O, Martins I, Ghiringhelli F, et al. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 2010;29:482–91. doi: 10.1038/onc.2009.356. [DOI] [PubMed] [Google Scholar]
- 26.Senovilla L, Vitale I, Martins I, Tailler M, Pailleret C, Michaud M, et al. An immunosurveillance mechanism controls cancer cell ploidy. Science. 2012;337:1678–84. doi: 10.1126/science.1224922. [DOI] [PubMed] [Google Scholar]
- 27.Garrido G, Rabasa A, Sánchez B, López MV, Blanco R, López A, et al. Induction of immunogenic apoptosis by blockade of epidermal growth factor receptor activation with a specific antibody. J Immunol. 2011;187:4954–66. doi: 10.4049/jimmunol.1003477. [DOI] [PubMed] [Google Scholar]
- 28.Sonnemann J, Gressmann S, Becker S, Wittig S, Schmudde M, Beck JF. The histone deacetylase inhibitor vorinostat induces calreticulin exposure in childhood brain tumour cells in vitro. Cancer Chemother Pharmacol. 2010;66:611–6. doi: 10.1007/s00280-010-1302-4. [DOI] [PubMed] [Google Scholar]
- 29.Molinari R, D’Eliseo D, Manzi L, Zolla L, Velotti F, Merendino N. The n3-polyunsaturated fatty acid docosahexaenoic acid induces immunogenic cell death in human cancer cell lines via pre-apoptotic calreticulin exposure. Cancer Immunol Immunother. 2011;60:1503–7. doi: 10.1007/s00262-011-1074-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Spisek R, Charalambous A, Mazumder A, Vesole DH, Jagannath S, Dhodapkar MV. Bortezomib enhances dendritic cell (DC)-mediated induction of immunity to human myeloma via exposure of cell surface heat shock protein 90 on dying tumor cells: therapeutic implications. Blood. 2007;109:4839–45. doi: 10.1182/blood-2006-10-054221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Miyamoto S, Inoue H, Nakamura T, Yamada M, Sakamoto C, Urata Y, et al. Coxsackievirus B3 is an oncolytic virus with immunostimulatory properties that is active against lung adenocarcinoma. Cancer Res. 2012;72:2609–21. doi: 10.1158/0008-5472.CAN-11-3185. [DOI] [PubMed] [Google Scholar]
- 32.Chen HM, Wang PH, Chen SS, Wen CC, Chen YH, Yang WC, et al. Shikonin induces immunogenic cell death in tumor cells and enhances dendritic cell-based cancer vaccine. Cancer Immunol Immunother. 2012;61:1989–2002. doi: 10.1007/s00262-012-1258-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Chen J, Xie J, Jiang Z, Wang B, Wang Y, Hu X. Shikonin and its analogs inhibit cancer cell glycolysis by targeting tumor pyruvate kinase-M2. Oncogene. 2011;30:4297–306. doi: 10.1038/onc.2011.137. [DOI] [PubMed] [Google Scholar]
- 34.Martins I, Kepp O, Schlemmer F, Adjemian S, Tailler M, Shen S, et al. Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress. Oncogene. 2011;30:1147–58. doi: 10.1038/onc.2010.500. [DOI] [PubMed] [Google Scholar]
- 35.Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2012;1:1111–34. doi: 10.4161/onci.21494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1:699–716. doi: 10.4161/onci.20696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology. 2012;1:306–15. doi: 10.4161/onci.19549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology. 2012;1:28–37. doi: 10.4161/onci.1.1.17938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Menger L, Vacchelli E, Kepp O, Eggermont A, Tartour E, Zitvogel L, et al. Trial Watch: Cardiac glycosides and cancer therapy. OncoImmunology. 2013;2 doi: 10.4161/onci.23082. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, et al. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology. 2012;1:1323–43. doi: 10.4161/onci.22009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Vacchelli E, Eggermont A, Galon J, Sautes-Fridman C, Zitvogel L, Kroemer G, et al. Trial Watch: Monoclonal antibodies in cancer therapy. OncoImmunology. 2013;2 doi: 10.4161/onci.22789. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1:894–907. doi: 10.4161/onci.20931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, et al. Trial Watch: Immunostimulatory cytokines. Oncoimmunology. 2012;1:493–506. doi: 10.4161/onci.20459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2012;1:179–88. doi: 10.4161/onci.1.2.19026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology. 2012;1:1557–76. doi: 10.4161/onci.22428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Garrido G, Sanchez B, Rodriguez HM, Lorenzano P, Alonso D, Fernandez LE. 7A7 MAb: a new tool for the pre-clinical evaluation of EGFR-based therapies. Hybrid Hybridomics. 2004;23:168–75. doi: 10.1089/1536859041224280. [DOI] [PubMed] [Google Scholar]
- 47.de La Motte Rouge T, Galluzzi L, Olaussen KA, Zermati Y, Tasdemir E, Robert T, et al. A novel epidermal growth factor receptor inhibitor promotes apoptosis in non-small cell lung cancer cells resistant to erlotinib. Cancer Res. 2007;67:6253–62. doi: 10.1158/0008-5472.CAN-07-0538. [DOI] [PubMed] [Google Scholar]
- 48.Boehrer S, Adès L, Braun T, Galluzzi L, Grosjean J, Fabre C, et al. Erlotinib exhibits antineoplastic off-target effects in AML and MDS: a preclinical study. Blood. 2008;111:2170–80. doi: 10.1182/blood-2007-07-100362. [DOI] [PubMed] [Google Scholar]
- 49.Goldberg RM. Cetuximab. Nat Rev Drug Discov. 2005;(Suppl):S10–1. doi: 10.1038/nrd1728. [DOI] [PubMed] [Google Scholar]
- 50.Pollack BP. EGFR inhibitors, MHC expression and immune responses : Can EGFR inhibitors be used as immune response modifiers? Oncoimmunology. 2012;1:71–4. doi: 10.4161/onci.1.1.18073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kummar S, Ji J, Morgan R, Lenz HJ, Puhalla SL, Belani CP, et al. A phase I study of veliparib in combination with metronomic cyclophosphamide in adults with refractory solid tumors and lymphomas. Clin Cancer Res. 2012;18:1726–34. doi: 10.1158/1078-0432.CCR-11-2821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Navid F, Baker SD, McCarville MB, Stewart CF, Billups CA, Wu J, et al. Phase I and clinical pharmacology study of bevacizumab, sorafenib, and low-dose cyclophosphamide in children and young adults with refractory/recurrent solid tumors. Clin Cancer Res. 2013;19:236–46. doi: 10.1158/1078-0432.CCR-12-1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ellis LM. Bevacizumab. Nat Rev Drug Discov. 2005;(Suppl):S8–9. doi: 10.1038/nrd1727. [DOI] [PubMed] [Google Scholar]
- 54.Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, Smith RA, et al. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discov. 2006;5:835–44. doi: 10.1038/nrd2130. [DOI] [PubMed] [Google Scholar]
- 55.Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med. 2012 doi: 10.1038/nm.2883. In press. [DOI] [PubMed] [Google Scholar]
- 56.Luke JJ, D’Adamo DR, Dickson MA, Keohan ML, Carvajal RD, Maki RG, et al. The cyclin-dependent kinase inhibitor flavopiridol potentiates doxorubicin efficacy in advanced sarcomas: preclinical investigations and results of a phase I dose-escalation clinical trial. Clin Cancer Res. 2012;18:2638–47. doi: 10.1158/1078-0432.CCR-11-3203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, et al. Global ARCC Trial Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007;356:2271–81. doi: 10.1056/NEJMoa066838. [DOI] [PubMed] [Google Scholar]
- 58.Moroney J, Fu S, Moulder S, Falchook G, Helgason T, Levenback C, et al. Phase I study of the antiangiogenic antibody bevacizumab and the mTOR/hypoxia-inducible factor inhibitor temsirolimus combined with liposomal doxorubicin: tolerance and biological activity. Clin Cancer Res. 2012;18:5796–805. doi: 10.1158/1078-0432.CCR-12-1158. [DOI] [PubMed] [Google Scholar]
- 59.Mamot C, Ritschard R, Wicki A, Stehle G, Dieterle T, Bubendorf L, et al. Tolerability, safety, pharmacokinetics, and efficacy of doxorubicin-loaded anti-EGFR immunoliposomes in advanced solid tumours: a phase 1 dose-escalation study. Lancet Oncol. 2012;13:1234–41. doi: 10.1016/S1470-2045(12)70476-X. [DOI] [PubMed] [Google Scholar]
- 60.Fassnacht M, Terzolo M, Allolio B, Baudin E, Haak H, Berruti A, et al. FIRM-ACT Study Group Combination chemotherapy in advanced adrenocortical carcinoma. N Engl J Med. 2012;366:2189–97. doi: 10.1056/NEJMoa1200966. [DOI] [PubMed] [Google Scholar]
- 61.Kim R. FOLFIRINOX: a new standard treatment for advanced pancreatic cancer? Lancet Oncol. 2011;12:8–9. doi: 10.1016/S1470-2045(10)70237-0. [DOI] [PubMed] [Google Scholar]
- 62.Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, et al. Groupe Tumeurs Digestives of Unicancer. PRODIGE Intergroup FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–25. doi: 10.1056/NEJMoa1011923. [DOI] [PubMed] [Google Scholar]
- 63.Gourgou-Bourgade S, Bascoul-Mollevi C, Desseigne F, Ychou M, Bouché O, Guimbaud R, et al. Impact of FOLFIRINOX Compared With Gemcitabine on Quality of Life in Patients With Metastatic Pancreatic Cancer: Results From the PRODIGE 4/ACCORD 11 Randomized Trial. J Clin Oncol. 2013;31:23–9. doi: 10.1200/JCO.2012.44.4869. [DOI] [PubMed] [Google Scholar]
- 64.Mondo EL, Noel MS, Katz AW, Schoeniger LO, Hezel AF. Unresectable locally advanced pancreatic cancer: Treatment with neoadjuvant leucovorin, fluorouracil, irinotecan, and oxaliplatin and assessment of surgical resectability. J Clin Oncol. 2013;31:e37–9. doi: 10.1200/JCO.2012.44.0339. [DOI] [PubMed] [Google Scholar]
- 65.Bang YJ, Kim YW, Yang HK, Chung HC, Park YK, Lee KH, et al. CLASSIC trial investigators Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): a phase 3 open-label, randomised controlled trial. Lancet. 2012;379:315–21. doi: 10.1016/S0140-6736(11)61873-4. [DOI] [PubMed] [Google Scholar]
- 66.Lee J, Park SH, Chang HM, Kim JS, Choi HJ, Lee MA, et al. Gemcitabine and oxaliplatin with or without erlotinib in advanced biliary-tract cancer: a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2012;13:181–8. doi: 10.1016/S1470-2045(11)70301-1. [DOI] [PubMed] [Google Scholar]
- 67.Colombo N, Kutarska E, Dimopoulos M, Bae DS, Rzepka-Gorska I, Bidzinski M, et al. Randomized, open-label, phase III study comparing patupilone (EPO906) with pegylated liposomal doxorubicin in platinum-refractory or -resistant patients with recurrent epithelial ovarian, primary fallopian tube, or primary peritoneal cancer. J Clin Oncol. 2012;30:3841–7. doi: 10.1200/JCO.2011.38.8082. [DOI] [PubMed] [Google Scholar]
- 68.Qayum N, Im J, Stratford MR, Bernhard EJ, McKenna WG, Muschel RJ. Modulation of the tumor microvasculature by phosphoinositide-3 kinase inhibition increases doxorubicin delivery in vivo. Clin Cancer Res. 2012;18:161–9. doi: 10.1158/1078-0432.CCR-11-1413. [DOI] [PubMed] [Google Scholar]
- 69.Zhang S, Liu X, Bawa-Khalfe T, Lu LS, Lyu YL, Liu LF, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med. 2012;18:1639–42. doi: 10.1038/nm.2919. [DOI] [PubMed] [Google Scholar]
- 70.Sondak VK, Smalley KS, Kudchadkar R, Grippon S, Kirkpatrick P. Ipilimumab. Nat Rev Drug Discov. 2011;10:411–2. doi: 10.1038/nrd3463. [DOI] [PubMed] [Google Scholar]
- 71.Wilmott JS, Scolyer RA, Long GV, Hersey P. Combined targeted therapy and immunotherapy in the treatment of advanced melanoma. Oncoimmunology. 2012;1:997–9. doi: 10.4161/onci.19865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Sabatini DM. mTOR and cancer: insights into a complex relationship. Nat Rev Cancer. 2006;6:729–34. doi: 10.1038/nrc1974. [DOI] [PubMed] [Google Scholar]
- 73.Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol. 2012;31:71–5. doi: 10.1038/nbt.2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Hassan R, Ho M. Mesothelin targeted cancer immunotherapy. Eur J Cancer. 2008;44:46–53. doi: 10.1016/j.ejca.2007.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Wang K, Bodempudi V, Liu Z, Borrego-Diaz E, Yamoutpoor F, Meyer A, et al. Inhibition of mesothelin as a novel strategy for targeting cancer cells. PLoS One. 2012;7:e33214. doi: 10.1371/journal.pone.0033214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, et al. Molecular mechanisms of cisplatin resistance. Oncogene. 2012;31:1869–83. doi: 10.1038/onc.2011.384. [DOI] [PubMed] [Google Scholar]
- 77.Tajeddine N, Galluzzi L, Kepp O, Hangen E, Morselli E, Senovilla L, et al. Hierarchical involvement of Bak, VDAC1 and Bax in cisplatin-induced cell death. Oncogene. 2008;27:4221–32. doi: 10.1038/onc.2008.63. [DOI] [PubMed] [Google Scholar]
- 78.Liu X, Shi Y, Maag DX, Palma JP, Patterson MJ, Ellis PA, et al. Iniparib nonselectively modifies cysteine-containing proteins in tumor cells and is not a bona fide PARP inhibitor. Clin Cancer Res. 2012;18:510–23. doi: 10.1158/1078-0432.CCR-11-1973. [DOI] [PubMed] [Google Scholar]
- 79.Patel AG, De Lorenzo SB, Flatten KS, Poirier GG, Kaufmann SH. Failure of iniparib to inhibit poly(ADP-Ribose) polymerase in vitro. Clin Cancer Res. 2012;18:1655–62. doi: 10.1158/1078-0432.CCR-11-2890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Friedberg JW. Rituximab for early-stage diffuse large-B-cell lymphoma. Lancet Oncol. 2006;7:357–9. doi: 10.1016/S1470-2045(06)70668-4. [DOI] [PubMed] [Google Scholar]
- 81.Gajraj E, Chung H, Longson C, Stevens A. Rituximab for follicular non-Hodgkin lymphoma. Lancet Oncol. 2008;9:320–1. doi: 10.1016/S1470-2045(08)70036-6. [DOI] [PubMed] [Google Scholar]
- 82.Mayor S. MET inhibitors: translating from bench to bedside. Lancet Oncol. 2011;12:14. doi: 10.1016/S1470-2045(10)70302-8. [DOI] [PubMed] [Google Scholar]
- 83.Stone RM. How I treat patients with myelodysplastic syndromes. Blood. 2009;113:6296–303. doi: 10.1182/blood-2008-09-038935. [DOI] [PubMed] [Google Scholar]
- 84.Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11:215–33. doi: 10.1038/nrd3626. [DOI] [PubMed] [Google Scholar]
- 85.Malvicini M, Alaniz L, Bayo J, Garcia M, Piccioni F, Fiore E, et al. Single low-dose cyclophosphamide combined with interleukin-12 gene therapy is superior to a metronomic schedule in inducing immunity against colorectal carcinoma in mice. Oncoimmunology. 2012;1:1038–47. doi: 10.4161/onci.20684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Shultz LD, Brehm MA, Garcia-Martinez JV, Greiner DL. Humanized mice for immune system investigation: progress, promise and challenges. Nat Rev Immunol. 2012;12:786–98. doi: 10.1038/nri3311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bollag DM, McQueney PA, Zhu J, Hensens O, Koupal L, Liesch J, et al. Epothilones, a new class of microtubule-stabilizing agents with a taxol-like mechanism of action. Cancer Res. 1995;55:2325–33. [PubMed] [Google Scholar]
- 88.Kowalski RJ, Giannakakou P, Hamel E. Activities of the microtubule-stabilizing agents epothilones A and B with purified tubulin and in cells resistant to paclitaxel (Taxol(R)) J Biol Chem. 1997;272:2534–41. doi: 10.1074/jbc.272.4.2534. [DOI] [PubMed] [Google Scholar]
- 89.Nicolaou KC, Winssinger N, Pastor J, Ninkovic S, Sarabia F, He Y, et al. Synthesis of epothilones A and B in solid and solution phase. Nature. 1997;387:268–72. doi: 10.1038/387268a0. [DOI] [PubMed] [Google Scholar]
- 90.Cowden CJ, Paterson I. Synthetic chemistry. Cancer drugs better than taxol? Nature. 1997;387:238–9. doi: 10.1038/387238a0. [DOI] [PubMed] [Google Scholar]
- 91.Service RF. Tumor-killer made; how does it work? Science. 1996;274:2009. doi: 10.1126/science.274.5295.2009. [DOI] [PubMed] [Google Scholar]
- 92.Burtness BA, Manola J, Axelrod R, Argiris A, Forastiere AA, Eastern Cooperative Oncology Group A randomized phase II study of ixabepilone (BMS-247550) given daily x 5 days every 3 weeks or weekly in patients with metastatic or recurrent squamous cell cancer of the head and neck: an Eastern Cooperative Oncology Group study. Ann Oncol. 2008;19:977–83. doi: 10.1093/annonc/mdm591. [DOI] [PubMed] [Google Scholar]
- 93.Galsky MD, Small EJ, Oh WK, Chen I, Smith DC, Colevas AD, et al. Multi-institutional randomized phase II trial of the epothilone B analog ixabepilone (BMS-247550) with or without estramustine phosphate in patients with progressive castrate metastatic prostate cancer. J Clin Oncol. 2005;23:1439–46. doi: 10.1200/JCO.2005.09.042. [DOI] [PubMed] [Google Scholar]
- 94.Rustin G, Reed N, Jayson GC, Ledermann JA, Adams M, Perren T, et al. A phase II trial evaluating two schedules of sagopilone (ZK-EPO), a novel epothilone, in patients with platinum-resistant ovarian cancer. Ann Oncol. 2011;22:2411–6. doi: 10.1093/annonc/mdq780. [DOI] [PubMed] [Google Scholar]
- 95.Vansteenkiste J, Lara PN, Jr., Le Chevalier T, Breton JL, Bonomi P, Sandler AB, et al. Phase II clinical trial of the epothilone B analog, ixabepilone, in patients with non small-cell lung cancer whose tumors have failed first-line platinum-based chemotherapy. J Clin Oncol. 2007;25:3448–55. doi: 10.1200/JCO.2006.09.7097. [DOI] [PubMed] [Google Scholar]
- 96.Bystricky B, Chau I. Patupilone in cancer treatment. Expert Opin Investig Drugs. 2011;20:107–17. doi: 10.1517/13543784.2011.542148. [DOI] [PubMed] [Google Scholar]
- 97.Kaklamani VG, Xu BH, Gomez HL. Global experience with ixabepilone in breast cancer. Expert Rev Anticancer Ther. 2011;11:683–92. doi: 10.1586/era.11.41. [DOI] [PubMed] [Google Scholar]