2,137
Views
97
CrossRef citations to date
0
Altmetric
Review

Trial watch

Chemotherapy with immunogenic cell death inducers

, , , , , & show all
Pages 179-188 | Published online: 01 Mar 2012

Abstract

The long-established notion that apoptosis would be immunologically silent, and hence it would go unnoticed by the immune system, if not tolerogenic, and hence it would actively suppress immune responses, has recently been revisited. In some instances, indeed, cancer cells undergo apoptosis while emitting a spatiotemporally-defined combination of signals that renders them capable of eliciting a long-term protective antitumor immune response. Importantly, only a few anticancer agents can stimulate such an immunogenic cell death. These include cyclophosphamide, doxorubicin and oxaliplatin, which are currently approved by FDA for the treatment of multiple hematologic and solid malignancies, as well as mitoxantrone, which is being used in cancer therapy and against multiple sclerosis. In this Trial Watch, we will review and discuss the progress of recent (initiated after January 2008) clinical trials evaluating the off-label use of cyclophosphamide, doxorubicin, oxaliplatin and mitoxantrone.

Introduction

For a long time, cell death instances were cataloged into either of two mutually exclusive and diametrically opposed categories: necrosis, constituting an accidental, pathological, morphologically nebulous and pro-inflammatory cell death subroutine, and apoptosis, being a finely regulated, physiological, morphologically defined and immunologically silent (if not tolerogenic) one.Citation1-Citation3 During the last decade, this textbook dichotomy has been revisited, if not invalidated. Thus, multiple pieces of evidence have accumulated to disconfirm the near-to-dogmatic notions that necrosis would constitute a merely accidental cell death mode,Citation4 that necrosis would fail to exhibit peculiar morphological manifestations,Citation5 that necrosis would not be involved in physiological processes,Citation5 and that apoptosis would always fail to elicit inflammatory and immune responses.Citation6

In 2005, Casares et al. first reported that tumor cells succumbing in vitro to anthracyclines, notably doxorubicin (but not to other chemotherapeutics such as mitomycin C), can acquire the capacity to elicit tumor-specific immune responses when inoculated in syngenic mice.Citation7 Such immune responses were found to efficiently protect mice against subsequent re-challenges with live cells of the same type, de facto resulting in long-term vaccination.Citation7 Since then, great efforts have been dedicated to the elucidation of the molecular and cellular mechanisms that underlie immunogenic cell death (ICD), leading to the discovery that ICD relies on the emission of a spatiotemporally-defined combination of signals by dying cells.Citation8 Such signals include (though perhaps are not limited to),Citation9 (1) the endoplasmic reticulum (ER) stress-elicited, caspase-dependent pre-apoptotic co-exposure of the ER chaperons calreticulin (CRT) and ERp57 on the outer leaflet of the plasma membrane;Citation10-Citation14 (2) the autophagy-dependent pre-apoptotic secretion of ATP;Citation15-Citation17 (3) the post-apoptotic release of the non-histone chromatin binding protein high mobility group box 1 (HMGB1)Citation18; and (4) the cell surface exposure or release of heat-shock proteins (HSPs) including HSP70 and HSP90.Citation19,Citation20 For ICD to be productive, i.e., to elicit a long-term protective anticancer immune response, each of these signals must be properly decoded by the immune system. Thus, by binding to a hitherto uncharacterized receptor on the surface of dendritic cells (DCs), CRT acts as an “eat-me” signal, thereby stimulating the DC-mediated uptake of apoptotic corpses (and hence tumor antigens).Citation10,Citation21 Extracellular ATP not only functions as a “find-me” signal, thereby stimulating the local recruitment of immune effector cells,Citation22 but also binds to purinergic P2RX7 receptors on the surface of these cells, thereby triggering the activation of the NLRP3 inflammasome.Citation17,Citation23 This is a critical step for the induction of antitumor immunity, as the inflammasome catalyzes the proteolytic maturation and secretion of interleukin-1β (IL-1β), a cytokine that is required for the adequate polarization of interferon γ (IFNγ)-producing CD8+ T cells.Citation17 By binding to Toll-like receptor 4 (TLR4) on DCs, HMGB1 engages a MYD88-mediated signaling cascade leading to increased tumor antigen processing and cross-presentation to T cells.Citation24 Along similar lines, the presence of HSPs on the surface of dying tumor cells or in their vicinity promotes the formation of tumor antigen-HSP complexes that are processed by DCs for T cell cross-priming more efficiently than tumor antigens alone.Citation25

So far, only a few anticancer agents have been shown to kill tumor cells while inducing all these phenomena in the correct spatiotemporal order, thus eliciting bona fide ICD. These include some types of radiotherapy and only four chemotherapeutics: the DNA alkylating compound cyclophosphamide, the anthracyclines doxorubicin and mitoxantrone and the platinum derivative oxaliplatin.Citation7,Citation10,Citation26-Citation28 Importantly, anticancer agents failing to elicit one (or more) of the abovementioned sine quibus non are intrinsically unable to induce ICD, a defect that, at least in some instances, can be restored by targeted pharmacological interventions. Thus, cisplatin (a platinum derivative structurally related to oxaliplatin) alone fails to elicit the pre-apoptotic exposure of CRT, yet becomes able to do so when combined with ER stressors such as thapsigargin, rendering cisplatin-induced cell death immunogenic.Citation29 Recently, the histone deacetylase inhibitor vorinostat has been found to trigger CRT exposure in childhood brain tumor cell lines, in vitro.Citation30 However, the true potential of vorinostat as an inducer of ICD remain unexplored, and actually contrasting reports can be found in the literature on its immunostimulatory vs. immunosuppressive effects.Citation31-Citation33

At present, cyclophosphamide, doxorubicin and oxaliplatin are approved by FDA for the treatment of multiple malignancies (). Mitoxantrone is mainly used for a cancer-unrelated indication, multiple sclerosis, even though FDA has also approved mitoxantrone-containing combination regimens for the treatment of acute leukemia, non-Hodgkin’s lymphoma, breast and prostate cancer (). In this Trial Watch, we will discuss the progress of recent (started after January 2008) clinical studies evaluating the efficacy—as off-label medications—of the only four chemotherapeutics that so far have been described as bona fide ICD inducers.

Table 1. Currently approved indications for immunogenic chemotherapy*

Cyclophosphamide

Cyclophosphamide is a DNA alkylating agent belonging to the family of nitrogen mustards.Citation34 Upon conversion into 4-hydroxycyclophosphamide by hepatic mixed function oxidases, cyclophosphamide becomes able to add an alkyl group (CnH2n+1) to the nitrogen atom at position 7 in the imidazole group of purine DNA bases, thereby acquiring cytotoxic properties.Citation34 Although initially conceived as a cancer-selective drug (due to a presumed cancer cell-specific mechanism of activation), cyclophosphamide has rapidly turned out to be cytotoxic for multiple cell types, including immune cells.Citation34 Owing to this pharmacodynamic profile, cyclophosphamide has soon entered the clinical practice not only as an anticancer agent for the treatment of some forms of lymphoma, leukemia and solid tumors (), but also for the therapy of non-neoplastic autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis and multiple sclerosis.Citation35 Still, recent data indicate that, at odds with the immunosuppressive properties of cyclophosphamide at high doses, metronomic cyclophosphamide regimens exert profound immunostimulatory effects,Citation36 for instance by selectively depleting or inhibiting FOXP3+ regulatory T cells (Tregs).Citation37 Such immunostimulatory properties appear to at least contribute to, if not entirely explain, the therapeutic success of cyclophosphamide as an anticancer agent.Citation34

In the last three years (2009–2011), a consistent number of trials aimed at testing the clinical benefits of cyclophosphamide in cancer patients has been terminated and the corresponding results published in high impact scientific journals (source http://www.ncbi.nlm.nih.gov/sites/entrez). In most cases, these clinical studies investigated—in oncological settings in which cyclophosphamide is approved by FDA—either how variations in dosage and/or schedule affect safety and efficacy or whether the combination between cyclophosphamide and other chemotherapeutics improves the therapeutic outcome.Citation38-Citation42 In addition, cyclophosphamide-based combination regimens have been tested in both low- and intermediate-risk rhabdomyosarcoma pediatric patients, in both cases providing no significant benefits as compared with the control arm.Citation43,Citation44

At present, there are around 530 open clinical trials, all phases confounded, that test the efficacy of cyclophosphamide in oncological indications (source www.clinicaltrials.gov). Approximately 430 (320–355 phase I-II, 75–110 phase III-IV) of these studies are performed in clinical scenarios that correspond or overlap with cyclophosphamide indications. In addition, cyclophosphamide is being evaluated, alone or in combination with other anticancer agents, in several off-label settings (). Around 10% of these latter trials are advanced ones (phase III-IV), including four studies that test cyclophosphamide-including combination regimens in brain cancer—notably atypical teratoid/rhabdoid tumors, choroid plexus tumors and ependymoma—patients (NCT01014767, NCT01096368, NCT00683319, NCT00653068), as well as two studies that investigate the combination of cyclophosphamide with cancer vaccines in non-small cell lung cancer (NSCLC) patients (NCT01015443, NCT01444118). A consistent fraction of early trials (phase I-II) is testing whether cyclophosphamide (most often in association with the nucleoside analog fludarabine or with immunostimulatory interventions such as the administration of IL-2, DC-based vaccines or autologous T cell transfers) is safe and efficient in melanoma patients (NCT01005745, NCT00863330, NCT01339663, NCT01106235, NCT01435499, NCT00683670, NCT00937625, NCT00871481, NCT00722098, NCT01455259, NCT00604136, NCT00846833, NCT00910650, NCT01319565, NCT01236573, NCT01468818, NCT01369875, NCT01369888, NCT01271907, NCT01118091). The combination between cyclophosphamide and granulocyte-macrophage colony-stimulating factor (GM-CSF)-based vaccines (including GVAX) in the context of pancreatic cancer is being investigated by many groups (NCT01468870, NCT01417000, NCT01088789, NCT00727441, NCT01021800). Along similar lines, cyclophosphamide is currently being tested together with immunostimulatory approaches (including anti-PD1 and anti-OX40 monoclonal antibodies)Citation45 against colorectal (NCT01064375, NCT00785122, NCT01462513, NCT00986518) and prostate cancer (NCT01303705, NCT01093183, NCT01420965, NCT01140373, NCT00753220). Besides these main trends, there are several other oncological indications for which the putative benefits of cyclophosphamide are being investigated in phase I-II clinical trials (), including (but not limited to) glioblastoma (NCT01403285, NCT01454596), myelodysplastic syndrome (NCT01255319, NCT00604201, NCT01174108), medulloblastoma (NCT00867178, NCT01356290), NSCLC (NCT00960115, NCT01159288), renal cancer (NCT01462214, NCT00923845), rhabdomyosarcoma (NCT01222715, NCT01055314) and thymoma (NCT01100944, NCT01025089).

Table 2. Main trends of clinical trials evaluating the effects of cyclophosphamide as an off-label medication for cancer patients*

Taken together, these observations clearly suggest that cyclophosphamide is the subject of an intense wave of clinical investigation, most often owing to its multifaceted immunostimulatory functions. Importantly, while many clinical studies demonstrate that metronomic cyclophosphamide leads to improved T cell effector functions,Citation36,Citation46-Citation48 its long-term clinical efficacy as well as its added value as compared with other strategies for depleting/inhibiting Tregs remain a matter of debate.Citation49-Citation52 The results from ongoing clinical trials will help to clarify this issue.

Doxorubicin

Doxorubicin (also known as adriamycin) is a natural compound belonging to the class of anthracycline antibiotics that functions as a DNA intercalating agent.Citation53 As such, it impedes the progression of topoisomerase II along DNA, de facto blocking the resolution of quaternary DNA structures (while inducing an accumulation of topoisomerase-introduced single-strand breaks) and hence inhibiting DNA replication (and to a lesser degree transcription).Citation54 Doxorubicin is associated with common and relatively mild side effects, including nausea, vomiting and alopecia, as well as with dose-limiting cardiotoxicity, presumably due to the generation of mitochondriotoxic reactive oxygen species (ROS) upon the interaction between doxorubicin and iron.Citation55 Still, doxorubicin (alone or in combination with other chemotherapeutics including cyclophosphamide) is widely used for the treatment of some forms of leukemia, Hodgkin’s lymphoma as well as a plethora of solid neoplasms, including (but not limited to) bladder, breast, stomach, lung, ovarian and thyroid cancer (). Recent preclinical results from several laboratories worldwide indicate that doxorubicin not only triggers ICD but stimulates various aspects of the immune response against cancer.Citation36

During the last triennium, only a few doxorubicin-based clinical trials have been reported in major scientific journals (source www.clinicaltrials.gov), focusing on the optimization of dosage and schedule for FDA-approved indications.Citation39,Citation40 Now, approximately 370 open clinical trials, all phases confounded, are investigating the efficacy of doxorubicin against various types of cancer (source www.clinicaltrials.gov). Among these, some 270 trials (190–220 phase I-II, 50–80 phase III-IV) are performed in settings that match or exhibit some degree of overlaps with the FDA-approved indications of doxorubicin. Moreover, doxorubicin is being tested, alone or combined with other chemotherapeutics, as an off-label medication in multiple clinical scenarios (). Indicatively, 20% of these latter trials are advanced ones, including a large fraction of studies in which doxorubicin is tested for the neoadjuvant treatment of liver cancer, either in combination with chemotherapy or coupled to maneuvers for maximizing efficacy such as transhepatic artery chemoembolization (TACE) (NCT01387932, NCT01332669, NCT00617981, NCT00936689, NCT01327521, NCT00807300, NCT01015833, NCT01324076, NCT01004978, NCT00980460). Other advanced trials explore the use of doxorubicin against lymphoma (NCT00854568, NCT00722137), multiple myeloma (NCT00734877, NCT00670631) and endometrial cancer (NCT00883116, NCT00698620). Along similar lines, a consistent fraction of doxorubicin-based early clinical trials () is performed on patients affected by liver cancer (often in association with TACE or the anti-angiogenic compound sorafenib) (NCT00988195, NCT01125020, NCT00844883, NCT00877071, NCT01259024, NCT01033578, NCT01116635, NCT01272557, NCT00855218, NCT00990860, NCT00919009, NCT01381211, NCT00857805, NCT01009801, NCT01281943, NCT01011010, NCT00949182, NCT00956930), multiple myeloma (near-to-always in combination with the proteasomal inhibitor bortezomib) (NCT00720174, NCT00750815, NCT01215344, NCT00617591, NCT01246063, NCT00985907, NCT00863174, NCT00849251, NCT01394354, NCT00706953, NCT01177683, NCT00724568, NCT00872521, NCT01160484, NCT01101594, NCT01365559, NCT00925821, NCT01255514. NCT01371227, NCT00744354, NCT00814541, NCT01078441. NCT00742404, NCT01055301, NCT01328236) and endometrial cancer (NCT01100359, NCT00739830). In addition, the safety and efficacy of doxorubicin (in most cases combined with other chemotherapeutics) are being investigated in diffuse large B-cell lymphoma (NCT01361191, NCT01087424), osteosarcoma (NCT01459484, NCT00691236, NCT01258634), thymoma (NCT01025089, NCT01100944) and urothelial cancer patients (NCT01093066, NCT00808639).

Table 3. Main trends of clinical trials evaluating the effects of doxorubicin as an off-label medication for cancer patients*

In spite of the fact that both cyclophosphamide and doxorubicin have been recently ascribed with a consistent immunostimulatory potential, in particular when employed in metronomic regimens,Citation36 only for the former this notion appears to translate into a clinical interest. The reasons underlying this trend remain unclear, yet may be related to toxicological and/or economical factors.

Oxaliplatin

Oxaliplatin is a third-generation platinum coordination complex originally developed in the 1990s in the attempt to circumvent the common resistance of tumors against first- (cisplatin) and second-generation (carboplatin) compounds.Citation56 The molecular mechanisms whereby platinum derivatives, especially oxaliplatin, exert cytotoxic effects against cancer cells are complex and surely overtake their capacity to generate DNA adducts and to activate the pro-apoptotic transcription factor p53.Citation57-Citation60 Although cisplatin-refractory neoplasms are largely considered to be responsive to oxaliplatin, clinical data suggest that there may be some degree of cross-resistance.Citation61 Since 1996 in Europe and 2002 in the US, oxaliplatin is clinically employed for the treatment of colorectal cancer in combination with 5-fluorouracil and folinic acid (the FOLFOX protocol) (). Besides inducing ICD, oxaliplatin reportedly inhibits the expression of programmed death ligand 2 (PD-L2), thereby limiting immunosuppression by both DCs and tumor cells.Citation62

The triennium 2009–2011 has witnessed several high impact publications on the clinical profile of oxaliplatin (source www.clinicaltrials.gov). On one hand, several studies have evaluated dosage, schedule and the possible combination of oxaliplatin with other chemotherapeutics for the therapy of colorectal cancer.Citation63-Citation65 On the other hand, multiple reports suggest that oxaliplatin may constitute a valuable therapeutic option for a wide range of tumors, including NSCLC, pancreatic, biliary tract, gall bladder and ampullary cancer.Citation66-Citation72

Not less than 330 ongoing clinical trials, all phases confounded, currently investigate the efficacy of oxaliplatin in cancer patients (source www.clinicaltrials.gov). Approximately 190 (140–160 phase I-II, 30–50 phase III-IV) of these studies involve colorectal cancer patients. In addition, there are some 140 clinical trials that evaluate the potential of oxaliplatin, alone or combined with other interventions, as an off-label medication (). Among 12 advanced clinical trials, three investigate the efficacy of oxaliplatin plus gemcitabine (a nucleoside analog) in biliary tract cancer patients (NCT01313377, NCT01470443, NCT01149122). In addition, there are four phase III trials evaluating the benefits of oxaliplatin (combined with the nucleoside analog 5-fluorouracil or with its pro-drug capecitabine) against gastric cancer (NCT01470742, NCT00941655, NCT00718354, NCT00680901), and three phase III trials in which oxaliplatin (associated to nucleoside analogs) is tested in pancreatic cancer patients (NCT01314027, NCT01362582, NCT01121848). This trend is fully reflected in early clinical studies, including 10 trials on biliary tract cancer patients (NCT01267344, NCT01180153, NCT01234051, NCT01127555, NCT00881504, NCT01389414, NCT01247337, NCT00713687, NCT00779454, NCT01206049), 50 studies on esophageal and gastric cancer patients (NCT00711243, NCT01307956, NCT01191697, NCT00982592, NCT01246960, NCT01386346, NCT01443065, NCT00849615, NCT01333033, NCT01295086, NCT01362127, NCT01262482, NCT00673673, NCT00667420, NCT00816543, NCT00861094, NCT01467921, NCT01248299, NCT00711412, NCT00985556, NCT00961077, NCT01471132, NCT01364493, NCT01138904, NCT00952003, NCT01359397, NCT01396707, NCT01049620, NCT01216644, NCT01090505, NCT01331928, NCT01130805, NCT01129310, NCT00767377, NCT01364376, NCT00733616, NCT01283204, NCT01130337, NCT01070290, NCT01106066, NCT01160419, NCT01100801, NCT01197118, NCT01351038, NCT01472029, NCT01206218, NCT00854854, NCT01208103, NCT01202409, NCT01422993). In addition, oxaliplatin is being investigated (most often in combination with 5-fluorouracil) in 14 distinct clinical trials enrolling pancreatic cancer patients (NCT00690300, NCT01473303, NCT00707278, NCT01413022, NCT01397019, NCT01359007, NCT00786058, NCT01446458, NCT01063192, NCT00728000, NCT01394120, NCT01415713, NCT01209962, NCT01454180) as well as in eight studies based on prostate cancer patients (NCT01338792, NCT00871169, NCT01042028, NCT00636883, NCT00602602, NCT01048320, NCT00609336, NCT01383538).

Table 4. Main trends of clinical trials evaluating the effects of oxaliplatin as an off-label medication for cancer patients*

Thus, there appears to be a consistent interest in the clinical properties of oxaliplatin, in particular in the context of combination regimens that include nucleoside analogs. Results from recently terminated clinical trials suggest that oxaliplatin may be beneficial for a wide range of solid tumors. Although it is too early to discern whether this depends or not on the ability of oxaliplatin to trigger ICD, the oncological indications for which oxaliplatin is approved by FDA may soon increase.

Mitoxantrone

Similar to doxorubicin, mitoxantrone (a synthetic anthracenedione first developed in the mid 1980s) operates as an intercalating agent and inhibits topoisomerase II, thus impairing DNA replication, transcription and repair.Citation73 Mitoxantrone shares doxorubicin’s spectrum of adverse reactions, including immunosuppression and a dose-limiting cardiotoxicity that can develop during treatment as well as years after discontinuation.Citation74 Due to its immunosuppressive properties, mitoxantrone is successfully used in the clinic to limit the frequency of relapse and slow the progression of several variants of multiple sclerosis.Citation75 In addition, the FDA has approved mitoxantrone, alone or in combination with other chemotherapeutics or prednisone, for the therapy of acute leukemia, non-Hodgkin’s lymphoma, breast and prostate cancer ().

Results from recently terminated (2009–2011), randomized clinical trials confirm that mitoxantrone provides clinical benefits in children with acute lymphoblastic leukemia but suggest that cabazitaxel plus prednisone may be superior than mitoxantrone plus prednisone for the treatment of metastatic castration-resistant prostate cancer that progresses upon docetaxel-based therapy.Citation76,Citation77

Now, 36 open clinical trials are studying the efficacy of mitoxantrone against distinct types of cancer (source www.clinicaltrials.gov). Of these, 29 (23 phase I-II, 6 phase III-IV) are performed in settings that match mitoxantrone FDA-approved indications. Moreover, the safety and efficacy of mitoxantrone, alone or combined with other anticancer agents, are being evaluated in a few off-label scenarios (). Among these latter studies, one single advanced (phase III) trial is testing the association between mitoxantrone (or other chemotherapeutics) and the anti-CD20 monoclonal antibody rituximab in follicular lymphoma patients (NCT00774826). Along similar lines, mitoxantrone is currently being investigated in five early clinical trials for its efficacy against various types of lymphoma and T-cell prolymphocytic leukemia (NCT00901927, NCT01133158, NCT01186640, NCT01144403, NCT00712582).

Table 5. Main trends of clinical trials evaluating the efficacy of mitoxantrone as an off-label medication for cancer patients*

The reasons whereby mitoxantrone—at odds with doxorubicin, which is also an anthracycline—is not the subject of an intense wave of clinical studies as an off label medication remain unclear. Perhaps, this may be due to the fact that while doxorubicin-based metronomic regimens have already been developed (and shown not only to be devoid of immunosuppressive effects but also to actively stimulate immunity),Citation36 the same does not hold true for mitoxantrone, whose preferential toxicity for immune cells de facto underlies its anticancer potential against lymphoma and leukemia.

Concluding Remarks

For a long time, the immune system has been viewed as a rather passive bystander of cancer, so much that even the National Cancer Institute recommended testing the antineoplastic potential of new molecules in immunodeficient murine models. Now, it has become clear that the immune system plays a critical role not only during early oncogenesis, by keeping under surveillance transformed and potentially tumorigenic cells, but also during the response of established malignancies to therapy.Citation36,Citation78-Citation80 On one hand, indicators of an ongoing immune response, such as the extent or the composition of the intratumoral infiltrate, as well as polymorphisms in genes that code for immune modulators have been correlated with the outcome of therapy. On the other hand, several anticancer compounds (be they conventional chemotherapeutics or targeted agents) have recently been shown to stimulate antitumor immunity.Citation36

Among several molecular and cellular circuitries whereby anticancer agents can trigger tumor-specific immune responses stands the induction of ICD, a functionally peculiar type of apoptosis that is associated with a spatiotemporally defined combination of immunogenic signals.Citation8 In spite of the fact that the existence of ICD has been acknowledged only a few years ago,Citation10 some agents that are capable of triggering ICD have already been identified, including cyclophosphamide, doxorubicin, oxaliplatin and mitoxantrone. All these compounds are approved by FDA for cancer therapy, have been successfully used in the clinic for several years, and are now being investigated for their utility in a range of off-label applications. It is tempting to speculate—but cannot be formally demonstrated—that part of the clinical success of these chemicals is due to their ability to trigger ICD. Irrespective of these considerations, it will be interesting to see whether cyclophosphamide, doxorubicin, oxaliplatin and mitoxantrone will be approved for additional cancer-related indications, as well as if novel inducers of ICD will be identified and will make their way from the bench to the bedside. In addition, it remains to be seen whether ICD inducers may be advantageously combined with non-immunogenic conventional chemotherapeutics, targeted anticancer agents and/or immunostimulatory strategies.

Abbreviations:
CRT=

calreticulin

DCs=

dendritic cells

ER=

endoplasmic reticulum

GM-CF=

granulocyte-macrophage colony-stimulating factor

HMGB1=

high mobility group box 1

HSPs, heat-shock proteins=

ICD, immunogenic cell death

IFNγ=

interferon γ

IL=

interleukin

NSCLC=

non-small cell lung cancer

TACE=

transhepatic artery chemoembolization

TLR4=

Toll-like receptor 4

Tregs=

FOXP3+ regulatory T cells

Acknowledgments

Authors are supported by the Ligue contre le Cancer (équipes labelisées), AXA Chair for Longevity Research, Cancéropôle Ile-de-France, Institut National du Cancer (INCa), Fondation Bettencourt-Schueller, Fondation de France, Fondation pour la Recherche Médicale, Agence National de la Recherche, the European Commission (Apo-Sys, ArtForce, ChemoRes. Death-Train) and the LabEx Immuno-Oncology.

References

  • 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; http://dx.doi.org/10.1038/sj.cdd.4402148; PMID: 17431418
  • 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; http://dx.doi.org/10.1038/cdd.2011.96; PMID: 21760595
  • Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, et al. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ 2009; 16:3 - 11; http://dx.doi.org/10.1038/cdd.2008.150; PMID: 18846107
  • Galluzzi L, Vanden Berghe T, Vanlangenakker N, Buettner S, Eisenberg T, Vandenabeele P, et al. Programmed necrosis from molecules to health and disease. Int Rev Cell Mol Biol 2011; 289:1 - 35; http://dx.doi.org/10.1016/B978-0-12-386039-2.00001-8; PMID: 21749897
  • Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G. Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol 2010; 11:700 - 14; http://dx.doi.org/10.1038/nrm2970; PMID: 20823910
  • Green DR, Ferguson T, Zitvogel L, Kroemer G. Immunogenic and tolerogenic cell death. Nat Rev Immunol 2009; 9:353 - 63; http://dx.doi.org/10.1038/nri2545; PMID: 19365408
  • 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; http://dx.doi.org/10.1084/jem.20050915; PMID: 16365148
  • Zitvogel L, Kepp O, Kroemer G. Decoding cell death signals in inflammation and immunity. Cell 2010; 140:798 - 804; http://dx.doi.org/10.1016/j.cell.2010.02.015; PMID: 20303871
  • Kepp O, Galluzzi L, Martins I, Schlemmer F, Adjemian S, Michaud M, et al. Molecular determinants of immunogenic cell death elicited by anticancer chemotherapy. Cancer Metastasis Rev 2011; 30:61 - 9; http://dx.doi.org/10.1007/s10555-011-9273-4; PMID: 21249425
  • 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; http://dx.doi.org/10.1038/nm1523; PMID: 17187072
  • Panaretakis T, Joza N, Modjtahedi N, Tesniere A, Vitale I, Durchschlag M, et al. The co-translocation of ERp57 and calreticulin determines the immunogenicity of cell death. Cell Death Differ 2008; 15:1499 - 509; http://dx.doi.org/10.1038/cdd.2008.67; PMID: 18464797
  • 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; http://dx.doi.org/10.1038/emboj.2009.1; PMID: 19165151
  • 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; http://dx.doi.org/10.1158/0008-5472.CAN-11-0950; PMID: 21602432
  • Zappasodi R, Pupa SM, Ghedini GC, Bongarzone I, Magni M, Cabras AD, et al. Improved clinical outcome in indolent B-cell lymphoma patients vaccinated with autologous tumor cells experiencing immunogenic death. Cancer Res 2010; 70:9062 - 72; http://dx.doi.org/10.1158/0008-5472.CAN-10-1825; PMID: 20884630
  • 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; http://dx.doi.org/10.4161/cc.8.22.10026; PMID: 19855167
  • Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents. Science 2012; 334:1573 - 7; http://dx.doi.org/10.1126/science.1208347; PMID: 22174255
  • 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; http://dx.doi.org/10.1038/nm.2028; PMID: 19767732
  • Apetoh L, Ghiringhelli F, Tesniere A, Criollo A, Ortiz C, Lidereau R, et al. The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy. Immunol Rev 2007; 220:47 - 59; http://dx.doi.org/10.1111/j.1600-065X.2007.00573.x; PMID: 17979839
  • 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; http://dx.doi.org/10.1038/nm0598-581; PMID: 9585232
  • Srivastava P. Roles of heat-shock proteins in innate and adaptive immunity. Nat Rev Immunol 2002; 2:185 - 94; http://dx.doi.org/10.1038/nri749; PMID: 11913069
  • Chao MP, Jaiswal S, Weissman-Tsukamoto R, Alizadeh AA, Gentles AJ, Volkmer J, et al. Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci Transl Med 2010; 2:63ra94; http://dx.doi.org/10.1126/scitranslmed.3001375; PMID: 21178137
  • Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature 2009; 461:282 - 6; http://dx.doi.org/10.1038/nature08296; PMID: 19741708
  • Aymeric L, Apetoh L, Ghiringhelli F, Tesniere A, Martins I, Kroemer G, et al. Tumor cell death and ATP release prime dendritic cells and efficient anticancer immunity. Cancer Res 2010; 70:855 - 8; http://dx.doi.org/10.1158/0008-5472.CAN-09-3566; PMID: 20086177
  • 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; http://dx.doi.org/10.1038/nm1622; PMID: 17704786
  • 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 1999; 191:1957 - 64; http://dx.doi.org/10.1084/jem.191.11.1957; PMID: 10839810
  • Lee Y, Auh SL, Wang Y, Burnette B, Meng Y, Beckett M, et al. Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood 2009; 114:589 - 95; http://dx.doi.org/10.1182/blood-2009-02-206870; PMID: 19349616
  • 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; http://dx.doi.org/10.1158/0008-5472.CAN-10-2788; PMID: 21156650
  • 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; http://dx.doi.org/10.1038/onc.2009.356; PMID: 19881547
  • 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; http://dx.doi.org/10.1038/onc.2010.500; PMID: 21151176
  • 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; http://dx.doi.org/10.1007/s00280-010-1302-4; PMID: 20221600
  • Buglio D, Georgakis GV, Hanabuchi S, Arima K, Khaskhely NM, Liu YJ, et al. Vorinostat inhibits STAT6-mediated TH2 cytokine and TARC production and induces cell death in Hodgkin lymphoma cell lines. Blood 2008; 112:1424 - 33; http://dx.doi.org/10.1182/blood-2008-01-133769; PMID: 18541724
  • Lucas JL, Mirshahpanah P, Haas-Stapleton E, Asadullah K, Zollner TM, Numerof RP. Induction of Foxp3+ regulatory T cells with histone deacetylase inhibitors. Cell Immunol 2009; 257:97 - 104; http://dx.doi.org/10.1016/j.cellimm.2009.03.004; PMID: 19358983
  • Schmudde M, Friebe E, Sonnemann J, Beck JF, Broker BM. Histone deacetylase inhibitors prevent activation of tumour-reactive NK cells and T cells but do not interfere with their cytolytic effector functions. Cancer Lett 2010; 295:173 - 81; http://dx.doi.org/10.1016/j.canlet.2010.02.024; PMID: 20346580
  • Emadi A, Jones RJ, Brodsky RA. Cyclophosphamide and cancer: golden anniversary. Nat Rev Clin Oncol 2009; 6:638 - 47; http://dx.doi.org/10.1038/nrclinonc.2009.146; PMID: 19786984
  • Weiner HL, Cohen JA. Treatment of multiple sclerosis with cyclophosphamide: critical review of clinical and immunologic effects. Mult Scler 2002; 8:142 - 54; http://dx.doi.org/10.1191/1352458502ms790oa; PMID: 11990872
  • Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secrete ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 2012; In press
  • Nakahara T, Uchi H, Lesokhin AM, Avogadri F, Rizzuto GA, Hirschhorn-Cymerman D, et al. Cyclophosphamide enhances immunity by modulating the balance of dendritic cell subsets in lymphoid organs. Blood 2010; 115:4384 - 92; http://dx.doi.org/10.1182/blood-2009-11-251231; PMID: 20154220
  • Moebus V, Jackisch C, Lueck HJ, du Bois A, Thomssen C, Kurbacher C, et al. Intense dose-dense sequential chemotherapy with epirubicin, paclitaxel, and cyclophosphamide compared with conventionally scheduled chemotherapy in high-risk primary breast cancer: mature results of an AGO phase III study. J Clin Oncol 2010; 28:2874 - 80; http://dx.doi.org/10.1200/JCO.2009.24.7643; PMID: 20458045
  • Peyrade F, Jardin F, Thieblemont C, Thyss A, Emile JF, Castaigne S, et al. Attenuated immunochemotherapy regimen (R-miniCHOP) in elderly patients older than 80 years with diffuse large B-cell lymphoma: a multicentre, single-arm, phase 2 trial. Lancet Oncol 2011; 12:460 - 8; http://dx.doi.org/10.1016/S1470-2045(11)70069-9; PMID: 21482186
  • Burnell M, Levine MN, Chapman JA, Bramwell V, Gelmon K, Walley B, et al. Cyclophosphamide, epirubicin, and Fluorouracil versus dose-dense epirubicin and cyclophosphamide followed by Paclitaxel versus Doxorubicin and cyclophosphamide followed by Paclitaxel in node-positive or high-risk node-negative breast cancer. J Clin Oncol 2010; 28:77 - 82; http://dx.doi.org/10.1200/JCO.2009.22.1077; PMID: 19901117
  • Robak T, Jamroziak K, Gora-Tybor J, Stella-Holowiecka B, Konopka L, Ceglarek B, et al. Comparison of cladribine plus cyclophosphamide with fludarabine plus cyclophosphamide as first-line therapy for chronic lymphocytic leukemia: a phase III randomized study by the Polish Adult Leukemia Group (PALG-CLL3 Study). J Clin Oncol 2010; 28:1863 - 9; http://dx.doi.org/10.1200/JCO.2009.25.9630; PMID: 20212251
  • Robak T, Dmoszynska A, Solal-Celigny P, Warzocha K, Loscertales J, Catalano J, et al. Rituximab plus fludarabine and cyclophosphamide prolongs progression-free survival compared with fludarabine and cyclophosphamide alone in previously treated chronic lymphocytic leukemia. J Clin Oncol 2010; 28:1756 - 65; http://dx.doi.org/10.1200/JCO.2009.26.4556; PMID: 20194844
  • Arndt CA, Stoner JA, Hawkins DS, Rodeberg DA, Hayes-Jordan AA, Paidas CN, et al. Vincristine, actinomycin, and cyclophosphamide compared with vincristine, actinomycin, and cyclophosphamide alternating with vincristine, topotecan, and cyclophosphamide for intermediate-risk rhabdomyosarcoma: children's oncology group study D9803. J Clin Oncol 2009; 27:5182 - 8; http://dx.doi.org/10.1200/JCO.2009.22.3768; PMID: 19770373
  • Beverly Raney R, Walterhouse DO, Meza JL, Andrassy RJ, Breneman JC, Crist WM, et al. Results of the Intergroup Rhabdomyosarcoma Study Group D9602 protocol, using vincristine and dactinomycin with or without cyclophosphamide and radiation therapy, for newly diagnosed patients with low-risk embryonal rhabdomyosarcoma: a report from the Soft Tissue Sarcoma Committee of the Children's Oncology Group. J Clin Oncol 2011; 29:1312 - 8; http://dx.doi.org/10.1200/JCO.2010.30.4469; PMID: 21357783
  • Galluzzi L, Vacchelli E, Friedman WH, Galon J, Sautès-Friedman C, Tartour E, et al. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunol 2012; 1:28 - 37
  • Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F, et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother 2007; 56:641 - 8; http://dx.doi.org/10.1007/s00262-006-0225-8; PMID: 16960692
  • Greten TF, Ormandy LA, Fikuart A, Hochst B, Henschen S, Horning M, et al. Low-dose cyclophosphamide treatment impairs regulatory T cells and unmasks AFP-specific CD4+ T-cell responses in patients with advanced HCC. J Immunother 2010; 33:211 - 8; http://dx.doi.org/10.1097/CJI.0b013e3181bb499f; PMID: 20139774
  • Lord R, Nair S, Schache A, Spicer J, Somaihah N, Khoo V, et al. Low dose metronomic oral cyclophosphamide for hormone resistant prostate cancer: a phase II study. J Urol 2007; 177:2136 - 40, discussion 40; http://dx.doi.org/10.1016/j.juro.2007.01.143; PMID: 17509300
  • Chu CS, Boyer J, Schullery DS, Gimotty PA, Gamerman V, Bender J, et al. Phase I/II randomized trial of dendritic cell vaccination with or without cyclophosphamide for consolidation therapy of advanced ovarian cancer in first or second remission. Cancer Immunol Immunother 2011; In press http://dx.doi.org/10.1007/s00262-011-1081-8; PMID: 22021066
  • de Vries IJ, Castelli C, Huygens C, Jacobs JF, Stockis J, Schuler-Thurner B, et al. Frequency of circulating Tregs with demethylated FOXP3 intron 1 in melanoma patients receiving tumor vaccines and potentially Treg-depleting agents. Clin Cancer Res 2011; 17:841 - 8; http://dx.doi.org/10.1158/1078-0432.CCR-10-2227; PMID: 21177412
  • Pere H, Montier Y, Bayry J, Quintin-Colonna F, Merillon N, Dransart E, et al. A CCR4 antagonist combined with vaccines induces antigen-specific CD8+ T cells and tumor immunity against self antigens. Blood 2011; 118:4853 - 62; http://dx.doi.org/10.1182/blood-2011-01-329656; PMID: 21908423
  • Slingluff CL Jr., Petroni GR, Chianese-Bullock KA, Smolkin ME, Ross MI, Haas NB, et al. Randomized multicenter trial of the effects of melanoma-associated helper peptides and cyclophosphamide on the immunogenicity of a multipeptide melanoma vaccine. J Clin Oncol 2011; 29:2924 - 32; http://dx.doi.org/10.1200/JCO.2010.33.8053; PMID: 21690475
  • Weiss RB. The anthracyclines: will we ever find a better doxorubicin?. Semin Oncol 1992; 19:670 - 86; PMID: 1462166
  • Momparler RL, Karon M, Siegel SE, Avila F. Effect of adriamycin on DNA, RNA, and protein synthesis in cell-free systems and intact cells. Cancer Res 1976; 36:2891 - 5; PMID: 1277199
  • Hayek ER, Speakman E, Rehmus E. Acute doxorubicin cardiotoxicity. N Engl J Med 2005; 352:2456 - 7; http://dx.doi.org/10.1056/NEJM200506093522321; PMID: 15944435
  • Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, et al. Molecular mechanisms of cisplatin resistance. Oncogene 2011; In press http://dx.doi.org/10.1038/onc.2011.384; PMID: 21892204
  • Berndtsson M, Hagg M, Panaretakis T, Havelka AM, Shoshan MC, Linder S. Acute apoptosis by cisplatin requires induction of reactive oxygen species but is not associated with damage to nuclear DNA. Int J Cancer 2007; 120:175 - 80; http://dx.doi.org/10.1002/ijc.22132; PMID: 17044026
  • Gourdier I, Crabbe L, Andreau K, Pau B, Kroemer G. Oxaliplatin-induced mitochondrial apoptotic response of colon carcinoma cells does not require nuclear DNA. Oncogene 2004; 23:7449 - 57; http://dx.doi.org/10.1038/sj.onc.1208047; PMID: 15334062
  • 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; http://dx.doi.org/10.1038/onc.2008.63; PMID: 18362892
  • Galluzzi L, Morselli E, Kepp O, Tajeddine N, Kroemer G. Targeting p53 to mitochondria for cancer therapy. Cell Cycle 2008; 7:1949 - 55; http://dx.doi.org/10.4161/cc.7.13.6222; PMID: 18642442
  • Stordal B, Pavlakis N, Davey R. Oxaliplatin for the treatment of cisplatin-resistant cancer: a systematic review. Cancer Treat Rev 2007; 33:347 - 57; http://dx.doi.org/10.1016/j.ctrv.2007.01.009; PMID: 17383100
  • Lesterhuis WJ, Punt CJ, Hato SV, Eleveld-Trancikova D, Jansen BJ, Nierkens S, et al. Platinum-based drugs disrupt STAT6-mediated suppression of immune responses against cancer in humans and mice. J Clin Invest 2011; 121:3100 - 8; http://dx.doi.org/10.1172/JCI43656; PMID: 21765211
  • Ducreux M, Malka D, Mendiboure J, Etienne PL, Texereau P, Auby D, et al. Sequential versus combination chemotherapy for the treatment of advanced colorectal cancer (FFCD 2000-05): an open-label, randomised, phase 3 trial. Lancet Oncol 2011; 12:1032 - 44; http://dx.doi.org/10.1016/S1470-2045(11)70199-1; PMID: 21903473
  • Hecht JR, Trarbach T, Hainsworth JD, Major P, Jager E, Wolff RA, et al. Randomized, placebo-controlled, phase III study of first-line oxaliplatin-based chemotherapy plus PTK787/ZK 222584, an oral vascular endothelial growth factor receptor inhibitor, in patients with metastatic colorectal adenocarcinoma. J Clin Oncol 2011; 29:1997 - 2003; http://dx.doi.org/10.1200/JCO.2010.29.4496; PMID: 21464406
  • Moosmann N, von Weikersthal LF, Vehling-Kaiser U, Stauch M, Hass HG, Dietzfelbinger H, et al. Cetuximab plus capecitabine and irinotecan compared with cetuximab plus capecitabine and oxaliplatin as first-line treatment for patients with metastatic colorectal cancer: AIO KRK-0104–a randomized trial of the German AIO CRC study group. J Clin Oncol 2011; 29:1050 - 8; http://dx.doi.org/10.1200/JCO.2010.31.1936; PMID: 21300933
  • Raez LE, Kobina S, Santos ES. Oxaliplatin in first-line therapy for advanced non-small-cell lung cancer. Clin Lung Cancer 2010; 11:18 - 24; http://dx.doi.org/10.3816/CLC.2010.n.003; PMID: 20085863
  • Crane CH, Varadhachary GR, Yordy JS, Staerkel GA, Javle MM, Safran H, et al. Phase II trial of cetuximab, gemcitabine, and oxaliplatin followed by chemoradiation with cetuximab for locally advanced (T4) pancreatic adenocarcinoma: correlation of Smad4(Dpc4) immunostaining with pattern of disease progression. J Clin Oncol 2011; 29:3037 - 43; http://dx.doi.org/10.1200/JCO.2010.33.8038; PMID: 21709185
  • Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011; 364:1817 - 25; http://dx.doi.org/10.1056/NEJMoa1011923; PMID: 21561347
  • Zhu AX, Meyerhardt JA, Blaszkowsky LS, Kambadakone AR, Muzikansky A, Zheng H, et al. Efficacy and safety of gemcitabine, oxaliplatin, and bevacizumab in advanced biliary-tract cancers and correlation of changes in 18-fluorodeoxyglucose PET with clinical outcome: a phase 2 study. Lancet Oncol 2010; 11:48 - 54; http://dx.doi.org/10.1016/S1470-2045(09)70333-X; PMID: 19932054
  • Gruenberger B, Schueller J, Heubrandtner U, Wrba F, Tamandl D, Kaczirek K, et al. Cetuximab, gemcitabine, and oxaliplatin in patients with unresectable advanced or metastatic biliary tract cancer: a phase 2 study. Lancet Oncol 2010; 11:1142 - 8; http://dx.doi.org/10.1016/S1470-2045(10)70247-3; PMID: 21071270
  • Overman MJ, Varadhachary GR, Kopetz S, Adinin R, Lin E, Morris JS, et al. Phase II study of capecitabine and oxaliplatin for advanced adenocarcinoma of the small bowel and ampulla of Vater. J Clin Oncol 2009; 27:2598 - 603; http://dx.doi.org/10.1200/JCO.2008.19.7145; PMID: 19164203
  • Sharma A, Dwary AD, Mohanti BK, Deo SV, Pal S, Sreenivas V, et al. Best supportive care compared with chemotherapy for unresectable gall bladder cancer: a randomized controlled study. J Clin Oncol 2010; 28:4581 - 6; http://dx.doi.org/10.1200/JCO.2010.29.3605; PMID: 20855823
  • Fox ME, Smith PJ. Long-term inhibition of DNA synthesis and the persistence of trapped topoisomerase II complexes in determining the toxicity of the antitumor DNA intercalators mAMSA and mitoxantrone. Cancer Res 1990; 50:5813 - 8; PMID: 2168281
  • Stuart-Harris R, Pearson M, Smith IE, Olsen EG. Cardiotoxicity associated with mitoxantrone. Lancet 1984; 324:219 - 20; http://dx.doi.org/10.1016/S0140-6736(84)90498-7; PMID: 6146768
  • Fox EJ. Management of worsening multiple sclerosis with mitoxantrone: a review. Clin Ther 2006; 28:461 - 74; http://dx.doi.org/10.1016/j.clinthera.2006.04.013; PMID: 16750460
  • de Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JP, Kocak I, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet 2010; 376:1147 - 54; http://dx.doi.org/10.1016/S0140-6736(10)61389-X; PMID: 20888992
  • Parker C, Waters R, Leighton C, Hancock J, Sutton R, Moorman AV, et al. Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukaemia (ALL R3): an open-label randomised trial. Lancet 2010; 376:2009 - 17; http://dx.doi.org/10.1016/S0140-6736(10)62002-8; PMID: 21131038
  • Bindea G, Mlecnik B, Fridman WH, Galon J. The prognostic impact of anti-cancer immune response: a novel classification of cancer patients. Semin Immunopathol 2011; 33:335 - 40; http://dx.doi.org/10.1007/s00281-011-0264-x; PMID: 21461991
  • Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pages C, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006; 313:1960 - 4; http://dx.doi.org/10.1126/science.1129139; PMID: 17008531
  • Mlecnik B, Tosolini M, Kirilovsky A, Berger A, Bindea G, Meatchi T, et al. Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J Clin Oncol 2011; 29:610 - 8; http://dx.doi.org/10.1200/JCO.2010.30.5425; PMID: 21245428