2,965
Views
77
CrossRef citations to date
0
Altmetric
Review

Trial watch

Monoclonal antibodies in cancer therapy

, , , , , & show all
Article: e22789 | Received 05 Nov 2012, Accepted 05 Nov 2012, Published online: 01 Jan 2013

Abstract

During the past 20 years, dozens—if not hundreds—of monoclonal antibodies have been developed and characterized for their capacity to mediate antineoplastic effects, either as they activate/enhance tumor-specific immune responses, either as they interrupt cancer cell-intrinsic signal transduction cascades, either as they specifically delivery toxins to malignant cells or as they block the tumor-stroma interaction. Such an intense research effort has lead to the approval by FDA of no less than 14 distinct molecules for use in humans affected by hematological or solid malignancies. In the inaugural issue of OncoImmunology, we briefly described the scientific rationale behind the use of monoclonal antibodies in cancer therapy and discussed recent, ongoing clinical studies investigating the safety and efficacy of this approach in patients. Here, we summarize the latest developments in this exciting area of clinical research, focusing on high impact studies that have been published during the last 15 months and clinical trials launched in the same period to investigate the therapeutic profile of promising, yet hitherto investigational, monoclonal antibodies.

Introduction

In 1975, Georges Köhler and César Milstein described for the first time an experimental setup allowing for the cost-effective production of high quantities of antibodies exhibiting the same antigenic specificity.Citation1 This breakthrough discovery, which allowed Köhler and Milstein to win the 2004 Nobel Prize for Medicine or Physiology, de facto revolutionized an unbelievable number of research protocols and clinical applications.Citation2 During the past 30 years, the hybridoma technology originally described by Köhler and Milstein (which was purely murine and hence associated with relevant immunogenicity issues) has been consistently ameliorated, resulting in the production of ever more refined reagents for clinical applications.Citation3 In 1986, the US Food and Drud Administration (FDA) approved muromonab, a murine IgG1 specific for CD3, for the therapy of transplanted patients undergoing rejection.Citation4 Since then, dozens of monoclonal antibodies (mAbs), including murine, chimeric as well humanized reagents, have been approved by international regulatory agencies (including FDA and the European Medicines Agency, EMA) for use in humans against multiple diseases, including (but not limited to) autoimmune disorders and cancer.Citation2,Citation5 Of these, no less than 14 mAbs, including naked reagents as well as mAbs coupled to antibiotics or radioactive isotopes, are nowadays authorized for use in cancer patients ().Citation5,Citation6

Table 1. Monoclonal antibodies currently approved for cancer therapy*,**

One year ago, concomitant with the launch of OncoImmunology, we published the first Trial Watch of what has then become a monthly series,Citation6-Citation14 dealing with the use of mAbs in cancer therapy.Citation6 As described in details therein, mAbs that potentially exert antineoplastic effects belong to one of six, non-mutually exclusive classes: (1) mAbs that target cancer cell-intrinsic pro-survival signal transduction cascades (e.g., cetuximab, which inhibits the epidermal growth factor receptor, EGFR, and is currently employed for the therapy of colorectal cancer);Citation15 (2) mAbs that interrupt the trophic interaction between malignant cells and their stroma, hence indirectly inhibiting tumor growth (e.g., bevacizumab, which targets the vascular endothelial growth factor, VEGF, and is currently approved for use in patients affected by breast, lung, renal and colorectal carcinoma);Citation16 (3) mAbs that recognize antigens expressed on the surface of tumor cells and exert antineoplastic effects by engaging immune effector mechanisms such as antibody-dependent cellular cytotoxicity (ADCC),Citation5,Citation17,Citation18 antibody-dependent cellular phagocytosis (ADCP),Citation19 and complement-dependent cytotoxicity (CDC)Citation20,Citation21 (e.g., rituximab, a naked anti-CD20 currently approved for use in lymphoma patients);Citation22-Citation24 (4) trifunctional (bispecific) mAbs, which can bind two distinct antigenic targets while retaining the ability of activating immune effector functions (e.g., catumaxomab, an anti-CD3, anti-EpCAM chimeric mAb nowadays is use for the therapy of malignant ascites in patient with EpCAM+ neoplasms);Citation25 (5) immunoconjugates (e.g., 90Y-ibritumomab tiuxetan and 131I-tositumomab, two radionuclide-coupled anti-CD20 mAbs that are currently approved for the treatment of lymphoma patients);Citation26,Citation27 and (6) immunostimulatory mAbs, i.e., mAbs that facilitate the development of an antitumor immune response by influencing the balance between the immunogenicity of malignant cells and the immunosuppressive mechanisms that they normally establish (e.g., ipilimumab, an anti-CTLA-4 mAb nowadays employed in melanoma patients).Citation28-Citation31

Here, we will summarize the latest developments in the clinical development of mAbs for cancer therapy, focusing on high impact studies that have been published during the last 15 months and clinical trials that have been launched in the same period to investigate the therapeutic profile of promising, yet hitherto experimental, mAbs. Of note, in this period, FDA has authorized one novel mAb for use in cancer patients and one oncological indication for a mAb that had previously been approved for the therapy of non-oncological patients (source http://www.cancer.gov/cancertopics/druginfo). Thus, brentuximab vedotin, an anti-CD30 monomethyl auristatin E (MMAE) conjugate, is now approved for the treatment of relapsed Hodgkin lymphoma and relapsed systemic anaplastic large cell lymphoma patients,Citation32,Citation33 In addition, denosumab, a receptor activator of NFκB ligand (RANKL)-targeting mAb that was employed in postmenopausal women at risk for osteoporosis, can nowadays be used to increase bone mass in patients who are at high risk of fracture as they undergo androgen deprivation therapy for non-metastatic prostate cancer or adjuvant aromatase inhibitor therapy for breast cancer.Citation34 Finally, the approval status of rituximab and cetuximab has recently been expanded in terms of therapeutic schedules and therapy-eligible patients subsets, respectively, while the authorization of bevacizumab as a therapeutic option for breast cancer patients has been revoked ().

Update on Clinical Reports

Since the submission of our previous Trial Watch on this topic (August 2011),Citation6 high-impact journals have published the results of no less than 50 clinical studies investigating the safety and efficacy of mAbs for cancer therapy. Obviously, a majority of these studies involved FDA-approved mAbs (), including bevacizumab (16 studies),Citation35-Citation50 trastuzumab (11 studies),Citation46,Citation51-Citation60 rituximab (10 studies),Citation61-Citation70 cetuximab (8 studies),Citation71-Citation78 ipilimumab (4 studies),Citation79-Citation82 alemtuzumab (2 studies),Citation83,Citation84 and denosumab (1 study).Citation85 In addition, a few high-impact publications have reported on the clinical profile of experimental mAbs, including immunostimulatory anti-PD-1 and anti-PD-L1 mAbs,Citation86,Citation87 the anti-CD22 calecheamicin conjugate inotuzumab ozogamicin,Citation88 the anti-CD19 maytansinoid conjugate SAR3419,Citation89 cixutumumab and figitumumab, two distinct mAbs specific for insulin-like growth factor 1 receptor (IGF1R),Citation90,Citation91 pertuzumab, an anti-HER2 mAb,Citation92,Citation93 and the CS1-targeting mAb elotuzumab.Citation94,Citation95

Thus, the clinical potential of FDA-approved mAbs, most often combined with conventional chemotherapeutic approaches, is under intensive investigation, an effort that will presumably lead to the approval of further oncological indications for these agents in the near future. In this respect, bevacizumab has been found to prolong progression-free survival (PFS) and overall survival (OS) in ovarian cancer patients treated with carboplatin and paclitaxel, in two randomized Phase III studies,Citation41,Citation42 and to improve the rate of pathological complete responses (pCRs) among HER2-negative breast carcinoma patients undergoing neoadjuvant docetaxel-based chemotherapy, again in two large randomized Phase III trials.Citation44,Citation45 The clinical profile of trastuzumab has most often been evaluated in cohorts of breast cancer patients,Citation46,Citation51-Citation58,Citation60 reflecting the indications for which it has been authorized by FDA. In particular, two distinct, randomized Phase III studies demonstrated that the combination of neoadjuvant chemotherapy, trastuzumab and a chemical inhibitor of HER2, lapatinib, is clinically superior to use of either approach alone.Citation58,Citation60 Similar to trastuzumab, rituximab has most extensively been tested in clinical settings that match its FDA-approved indications, i.e., cohorts of chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL) patients.Citation61-Citation67 One notable exception to this trend is represented by the study of Kluin-Nelemans et al., demonstrating that rituximab in combination with cyclophosphamide, doxorubicin, vincristine, and prednisone is efficient in old patients affected by mantle-cell lymphoma.Citation70 Cetuximab has been tested in a few different clinical settings, including cohorts of non-small cell lung carcinoma (NSCLC), pancreatic cancer and colorectal carcinoma patients.Citation71-Citation78 Notably, EGFR expression levels have been shown to predict a survival benefit from the addition of cetuximab to first-line conventional chemotherapy in NSCLC patients.Citation74 Ipilimumab has been tested in cohorts of melanoma and NSCLC patients, alone or combined with paclitaxel and cisplatin, respectively,Citation81,Citation82 as well as in prostate cancer patients undergoing vaccination,Citation79,Citation80 invariably providing clinical benefits, at least to some extent. Finally, alemtuzumab and denosumab have been shown to improve the therapeutic potential of fludarabine in a cohort of CLL patients,Citation83 and to reduce the incidence of bone metastases in subjects affected by castration-resistant prostate carcinoma.Citation85

Some experimental mAbs have also generated remarkable clinical results during the last 15 months. For instance, the mAb-mediated blockade of the interaction between the inhibitory receptor PD-1 (which is expressed by T cells) and its ligand (PD-L1), undertaken as a standalone intervention with either anti-PD-1 (MDX-1106, BMS-936558) or anti-PD-L1 (MDX-1105, BMS-936559) mAbs, has been shown to induce tumor regression or disease stabilization in a relatively small (but encouraging) proportion of patients affected by NSCLC, melanoma and renal cell carcinoma (RCC).Citation86,Citation87 Inotuzumab ozogamicin has been associated with a promising therapeutic profile among refractory and relapsed acute lymphocytic leukemia patients.Citation88 The intravenous administration of SAR3419 to relapsed CD19+ B-cell lymphoma patients has been found to be safe and to elicit a consistent rate of clinical responses.Citation89 Along similar lines, anti-IGF1R mAbs such as cixutumumab and figitumumab have been shown to be well tolerated by children affected by refractory Ewing sarcoma, exhibiting modest antineoplastic activity as single agents.Citation90,Citation91 The inclusion of pertuzumab in combinatorial regimens involving trastuzumab (alone or combined with docetaxel-based chemotherapy) have been associated with improved PFS (compared with the use of either mAb alone or chemotherapy plus trastuzumab only), in cohorts of HER2+ breast cancer patients.Citation92,Citation93 Finally, in two distinct Phase I clinical trials, the combination of elotuzumab and bortezomib or lenalidomide plus low-dose dexamethasone was well tolerated and exhibited a promising efficacy in patients affected by relapsed or refractory multiple myeloma.Citation94,Citation95

Focusing on recent, high-impact mAb-related translational research, we have found of particular interest the studies by Yonesaka et al., Diaz et al. and Misale et al., demonstrating that colorectal carcinoma patients become refractory to EGFR-targeting mAbs including cetuximab and panitumumab along with the emergence of KRAS mutations or with an increased signaling via the EGFR-related receptor HER2;Citation96,Citation97 as well as the work by Prahallad et al., revealing that—at least in a fraction of cases—the unresponsiveness of colon carcinoma patients to BRAFV600E inhibitors stems from a feedback hyperactivation of EGFR signaling.Citation98

Update on Investigational Monoclonal Antibodies under Clinical Evaluation

When this Trial Watch was being redacted (October 2012), official sources listed 45 clinical trials launched after 2011, August 1 that would investigate the safety and therapeutic profile of hitherto investigational mAbs in cancer patients (source www.clinicaltrials.gov).

Bavituximab is a chimeric IgG1 specific for phosphatidylserine (PS), an anionic phospholipid that—under physiological conditions—is found in the inner leaflet of the plasma membrane.Citation99 PS translocates to the cell surface in some instances of cell death,Citation100-Citation102 cell activation and malignant transformation, and has been proposed to constitute a tumor vasculature-specific marker.Citation103,Citation104 Encouraging preclinical findings by Ran et al.Citation99 supported the evaluation of bavituximab in clinical settings. Recent results from a Phase I study indicate that bavituximab at doses up to 3 mg/Kg/week is well tolerated by patients with advanced solid tumors.Citation105 Recently (since 2011, August 1), one single Phase I trial has been launched to assess the tolerability and preliminary therapeutic profile of bavituximab, combined with capecitabine and radiotherapy, in rectal adenocarcinoma patients (NCT01634685) ().

Table 2. Clinical trials recently launched to evaluate the therapeutic profile of monoclonal antibodies*

BC8, a CD45-targeting mAb that most often is conjugated to radionuclides (e.g., 131I, 111In), is currently under preclinical and clinical investigation for the treatment of CD45-expressing hematological malignancies.Citation106 The combination of BC8 with cyclophosphamide and total body irradiation (TBI) has previously been shown to be tolerated and convey clinical benefits in a small cohort of patients affected by myelodysplastic syndrome (MDS) and acute lymphoid leukemia (ALL).Citation107 Recently (since 2011, August 1), two Phase I trials have been launched to test the clinical profile of BC8, one involving multiple myeloma patients treated with BC8 in combination with fludarabine and TBI (NCT01503242), and one involving patients with refractory or relapsed lymphoid malignancies receiving BC8 as a standalone intervention (NCT01678443), in both cases as a pre-transplantation conditioning regimen ().

Blinatumomab belongs to the category of so-called BiTEs (bispecific T-cell engagers), i.e., bi-specific mAbs that invariably target CD3 (a component of the TCR signal transduction machinery expressed by T cells) and a tumor-associated antigen (in this case, CD19, a transmembrane protein mainly expressed by B cells).Citation108 Hence, unlike conventional monospecific mAbs, blinatumomab exerts antineoplastic effects by physically bridging malignant B cells and host T cells, hence promoting the cytotoxic activity of the latter.Citation108 High response rates and durable remissions have been observed in the first clinical trials testing the safety and therapeutic profile of blinatumomab among B-cell NHL and B-precursor ALL patients.Citation109-Citation111 Recently (since 2011, August 1), two Phase I/II trials have been initiated to investigate the safety and efficacy of blinatumomab, given as a standalone intervention, in subjects affected by B-precursor ALL (NCT01466179, NCT01471782) ().

Ch14.18 is a chimeric IgG1 specific for GD2, a disialoganglioside GD2 that is often abundant at the surface neuroendocrine tumor cells.Citation112,Citation113 The evaluation of the safety and efficacy of Ch14.18 as a standalone agent for the treatment of melanoma and neuroblastoma has begun in the early 1990s,Citation114,Citation115 with relatively unsatisfactory results. Later on, a few clinical studies have investigated the clinical potential of combinatorial regimens consisting of Ch14.18 in association with immunostimulatory cytokines like interleukin (IL)-2 and granulocyte macrophage-colony stimulating factor (GM-CSF) or metronomic chemotherapy,Citation116-Citation119 reporting rather promising findings, in particular for the use of Ch14.18 in association with GM-CSF, IL-1 and isotretinoin (a retinoid) in high-risk neuroblastoma patients.Citation118 Recently (since 2011, August 1), two Phase I/II trials have been launched to test the therapeutic potential of Ch14.18, alone or combined with GM-CSF, IL-1 and isotretinoin, in neuroblastoma patients (NCT01418495; NCT01592045) ().

Cixutumumab (a fully human IgG1), dalotuzumab (a humanized IgG1) and ganitumab (a fully human IgG1) all target IGFR1, a transmembrane receptor that is overexpressed or hyperactivated by most, if not all, malignant tissues, hence operating as an anti-apoptotic signal transducer.Citation120 According to the results of early clinical trials, cixutumumab and dalotuzumab as single agents as well as the combination of cixutumumab and temsirolimus (an inhibitor of the intracellular signaling pathway elicited by IGFR1) are generally well tolerated by patients bearing advanced solid tumors,Citation91,Citation121-Citation123 with prominent adverse effects involving the dermis.Citation124 Conversely, dose-limiting toxicities have been reported to develop among unselected NSCLC patients treated with cixutumumab in combination with the EGFR inhibitor erlotinib at full dosage.Citation125 In 2012, results from 4 distinct clinical studies testing the safety and efficacy of ganitumab in patients affected by Ewing family tumors, pancreatic carcinoma or other solid malignancies have been published.Citation126-Citation129 Globally, ganitumab—both as a single agent and associated with targeted agents or conventional chemotherapy—appears to be well tolerated and to exert antineoplastic activity, at least in a fraction of patients.Citation126-Citation129 This said, results from less recent Phase III studies have shown that targeting the IGF1R pathway is not associated with clear clinical benefits in cancer patients.Citation130 Accordingly, several anti-IGF1R programs—including one large randomized Phase III study that originally aimed at testing the therapeutic potential of ganitumab among prostatic cancer patients (NCT01231347)—have lately been discontinued.Citation130,Citation131 Recently (since 2011, August 1), two Phase II clinical trials have been launched to test the clinical profile of cixutumumab, either as a standalone intervention or combined with temsirolimus, in melanoma, sarcoma and brain cancer patients (NCT01413191; NCT01614795). In addition, three Phase I/II studies have been initiated to investigate the safety and efficacy of dalotuzumab, alone or in combination with ridaforolimus (a functional analog of temsirolimus) or conventional chemotherapeutics, in subjects bearing breast carcinoma, colorectal carcinoma or advanced solid tumors (NCT01431547; NCT01605396; NCT01609231). Finally, four Phase I/II studies have been initiated to assess the therapeutic potential of ganitumab, in combination with conventional chemotherapy, radiotherapy or trastuzumab, in individuals affected by breast carcinoma, pancreatic carcinoma or other advanced solid neoplasms (NCT01298401; NCT01473303; NCT01479179; NCT01562899). Of note, one of these studies (NCT01473303) is listed as complete, though results are not available, while another one (NCT01479179) has been withdrawn prior to patient enrollment, which somehow reflects current feelings about mAb-based anti-IGF1R cancer therapy ().

MDX-1106 is a fully human IgG4 that specifically binds PD-1, a transmembrane receptor that—upon binding by its cognate ligand PD-L1, generates immunosuppressive signals in activated T cells.Citation132-Citation134 In 2012, MDX-1106 (as well as of the PD-L1-targeting antibody MDX-1105) has been shown to be well tolerated and to induce objective clinical responses in a relatively small (but encouraging) proportion of patients affected by advanced NSCLC, melanoma and RCC.Citation86,Citation87 Recently (since 2011, August 1), no less than 10 Phase I-III clinical trials have been launched to investigate the clinical potential of MDX-1105 and MDX-1106, given as standalone agents or combined with conventional chemotherapy (e.g., carboplatin + paclitaxel or cisplatin + gemcitabine or cisplatin + paclitaxel), targeted therapy (e.g., pazopanib or sunitinib, two tyrosine kinase receptor inhibitors) or immunostimulatory agents (e.g., recombinant IL-21), in cohorts of patients affected by hematological tumors (NCT01452334; NCT01592370), melanoma (NCT01621490), NSCLC (NCT01454102; NCT01642004; NCT01673867), hepatocellular carcinoma (NCT01658878), RCC (NCT01472081; NCT01668784), and advanced solid tumors (NCT01629758) (). Of note, the only study involving MDX-1105 (NCT01452334) has been withdrawn prior to patient enrollment, for undeclared reasons (source www.clinicaltrials.gov).

Both necitumumab (a fully human IgG1) and nimotuzumab (a humanized IgG1) specifically bind EGFR, a transmembrane receptor with tyrosine kinase activity that is hyperactivated, due to point mutations, in a number of solid malignancies. Accordingly, a number of EGFR-targeting therapeutics have already been approved by FDA and other regulatory agencies for use in cancer patients, including the cell-permeant tyrosine kinase inhibitors erlotinib and gefinitibCitation135-Citation137 and the anti-EGFR mAbs cetuximab and panitumumab ().Citation138,Citation139 Still, the clinical interest in EGFR-targeting approaches is not decreasing, at least in part owing to the fact that most, if not all, FDA approved EGFR-targeting agents have been associated with the development of chemoresistance in a consistent fraction of patients.Citation96-Citation98,Citation140 The clinical development of nimotuzumab appears to be more advanced than that of necitumumab. Indeed, during the last 15 months, several pilot or Phase I studies investigating the safety and preliminary therapeutic profile of nimotuzumab (given as a standalone intervention or combined with standard radio- or chemotherapeutic regimens) in patients affected by various solid malignancies have been concluded, overall reporting a low incidence of tolerable adverse effects and improved disease outcome, at least in a fraction of cases.Citation141-Citation154 Conversely, no reports have been published since 2010 on the clinical profile of necitumumab.Citation144 Recently (since 2011, August 1), a total of 9 Phase II clinical trials have been initiated to investigate the safety and efficacy of necitumumab (NCT01606748; NCT01624467) or nimotuzumab (NCT01425736; NCT01463605; NCT01486992; NCT01498562; NCT01516996; NCT01616849; NCT01688700), as single agents or associated with standard chemotherapy, in cohorts of head and neck cancer (NCT01425736, NCT01516996; NCT01616849) esophageal cancer (NCT01463605; NCT01486992; NCT01688700) and NSCLC (NCT01498562) patients, as well as of individuals affected by various solid tumors (NCT01606748; NCT01624467) ().

Ramucirumab (IMC-1121B) is a fully human IgG1 specific for the VEGF receptor 2 (VEGFR2). By antagonizing endogenous VEGF signaling,Citation155 ramucirumab inhibits of the most prominent interactions between neoplastic and stromal cells, i.e., neo-angiogenesis.Citation156 Early Phase I clinical trials demonstrated that ramucirumab is generally well tolerated by cancer patients,Citation155,Citation157 and prompted the launch of a randomized, double-blind Phase III study to evaluate whether ramucirumab can improve the efficacy of docetaxel-based chemotherapy in subjects bearing stage IV NSCLC (NCT01168973, registered at www.clinicaltrials.gov on July 2010).Citation158 This study has not yet been concluded. More recently (since 2011, August 1), 5 distinct Phase I clinical trials have been commenced to investigate the tolerability and efficacy of ramucirumab, most often combined with conventional chemotherapeutics, in subjects affected by breast carcinoma (NCT01427933) or other solid neoplasms (NCT01515306; NCT01567163; NCT01634555; NCT01682135) ().

Siltuximab (CNTO-328) is a chimeric IgG1 that binds to IL-6, a pleiotropic cytokine that reportedly modulates several aspects of oncogenesis and tumor progression, including the balance between cell proliferation and cell death as well as neo-angiogenesis.Citation159 Siltuximab-based monotherapy as well as combinatorial regimens including siltuximab and mitoxantrone + prednisone or siltuximab + docetaxel have lately been reported to be well tolerated by prostate cancer patients, although objective effects on disease outcome could not always be documented.Citation160-Citation162 Since 2011, August 1, 3 Phase I/II clinical trials have been launched to assess the therapeutic profile of siltuximab, as a single agent or combined with best supportive care or bortezomib + dexamethasone + lenalidomide, in MDS (NCT01513317) and multiple myeloma (NCT01484275; NCT01531998) patients (). Conversely, the Phase III trial NCT01266811, which was initiated in December 2010 to test the efficacy of siltuximab + bortezomib + dexamethasone in similar patient cohorts has been withdrawn prior to patient enrollment for undeclared reasons (source www.clinicaltrials.gov).

Tigatuzumab (CS-1008) is a humanized IgG1 that functions as an agonist for tumor necrosis factor-related apoptosis-inducing ligand receptor 2 (TRAILR2), hence promoting the death of TRAILR2-expressing malignant (but not non-transformed) cells.Citation163 Although the molecular basis underlying the selectivity of tigatuzumab remains unclear,Citation164,Citation165 the results of preliminary Phase I-II studies in cancer patients were encouraging, suggesting that tigatuzumab is generally well tolerated.Citation166 Recently (since 2011, August 1), 1 Phase II clinical trial has been initiated to test the safety and efficacy of tigatuzumab combined with abraxane (protein-bound paclitaxel particles), in breast carcinoma patients (NCT01307891) ().

Tremelimumab (ticilimumab, CP-675,206) is a human IgG2 that, similar to the FDA-approved mAb ipilimumab, inhibits the development of peripheral tolerance by antagonizing the immunosuppressive receptor CTLA-4 on the surface of T cells.Citation167,Citation168 Combinatorial regimens including tremelimumab + high-dose interferon α-2b (IFNα-2b) and tremelimumab + androgen deprivation have recently been shown to be well tolerated by melanoma and prostate carcinoma patients,Citation169,Citation170 respectively. Since 2011, August 1, 2 Phase II studies have been launched to investigate the clinical profile of tremelimumab, invariably employed as a standalone intervention, in mesothelioma patients (NCT01649024; NCT01655888) ().

Concluding Remarks

In no more than 15 months, besides extending the indications of mAbs that were previously approved in oncological settings, the US FSA has authorized the use in cancer patients of two novel mAbs, brentuximab vedotin and denosumab.Citation32-Citation34 In the very same period, (1) results from no less than 50 clinical studies testing the safety and antineoplastic profile of mAbs have been published in top-impact forums; and (2) approximately 50 novel mAb-based clinical trials enrolling cohorts of cancer patients have been registered at www.clinicaltrials.gov. Thus, the interest in the clinical development of mAbs for cancer therapy remains very high.

With two notable exceptions (i.e., NCT01266811 and NCT01231347), all Phase III studies that we listed in our latest Trial Watch dealing with mAbs and cancer therapy are still ongoing.Citation6 The results of these large, randomized clinical trials are likely to determine whether novel mAbs will soon be authorized by FDA and/or other international regulatory agencies for use in cancer patients. In addition, an intense wave of preclinical investigation is ongoing not only to discover novel mAb-druggable oncology-relevant targets, but also to generate further insights into the molecular and cellular circuitries whereby mAbs actually exert antineoplastic effects and how this knowledge can be translated into more stable and more efficient therapeutics.Citation6,Citation29,Citation171 In the mid-long term, such preclinical studies will surely generate the next generation of mAbs for clinical development.

Abbreviations:
ADCC =

antibody-dependent cellular cytotoxicity

ADCP =

antibody-dependent cellular phagocytosis

ALL =

acute lymphoid leukemia

BiTE =

bispecific T-cell engager

CDC =

complement-dependent cytotoxicity

CLL =

chronic lymphocytic leukemia

FDA =

Food and Drug Administration

EGFR =

epidermal growth factor receptor

EMA =

European Medicines Agency

GM-CSF =

granulocyte macrophage-colony stimulating factor

IFNα-2b =

interferon α-2b

IGF1R =

insulin-like growth factor 1 receptor

IL =

interleukin

mAb =

monoclonal antibody

MMAE =

monomethyl auristatin E

MDS =

myelodysplastic syndrome

NHL =

non-Hodgkin lymphoma

NSCLC =

non-small cell lung carcinoma

OS =

overall survival

pCR =

pathological complete response

PFS =

progression-free survival

PS =

phosphatidylserine

RANKL =

receptor activator of NFκB ligand

TRAILR2 =

tumor necrosis factor-related apoptosis-inducing ligand receptor 2

TBI =

total body irradiation

VEGF =

vascular endothelial growth factor

VEGFR2 =

VEGF receptor 2

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

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

  • Köhler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975; 256:495 - 7; http://dx.doi.org/10.1038/256495a0; PMID: 1172191
  • Alkan SS. Monoclonal antibodies: the story of a discovery that revolutionized science and medicine. Nat Rev Immunol 2004; 4:153 - 6; http://dx.doi.org/10.1038/nri1265; PMID: 15040588
  • Zou YR, Gu H, Rajewsky K. Generation of a mouse strain that produces immunoglobulin kappa chains with human constant regions. Science 1993; 262:1271 - 4; http://dx.doi.org/10.1126/science.8235658; PMID: 8235658
  • Ortho Multicenter Transplant Study Group. A randomized clinical trial of OKT3 monoclonal antibody for acute rejection of cadaveric renal transplants. N Engl J Med 1985; 313:337 - 42; http://dx.doi.org/10.1056/NEJM198508083130601; PMID: 2861567
  • Weiner LM, Surana R, Wang S. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat Rev Immunol 2010; 10:317 - 27; http://dx.doi.org/10.1038/nri2744; PMID: 20414205
  • 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; http://dx.doi.org/10.4161/onci.1.1.17938; PMID: 22720209
  • 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; http://dx.doi.org/10.4161/onci.19549; PMID: 22737606
  • 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; http://dx.doi.org/10.4161/onci.1.2.19026; PMID: 22720239
  • 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; http://dx.doi.org/10.4161/onci.21494
  • 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; http://dx.doi.org/10.4161/onci.20696; PMID: 22934262
  • 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; http://dx.doi.org/10.4161/onci.20931
  • Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, et al. Trial Watch: Immunostimulatory cytokines. Oncoimmunology 2012; 1:493 - 506; http://dx.doi.org/10.4161/onci.20459; PMID: 22754768
  • Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautes-Fridman C, et al. Trial Watch: Peptide vaccines in cancer therapy. OncoImmunology 2012; 1; In press http://dx.doi.org/10.4161/onci.22428
  • 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; In press http://dx.doi.org/10.4161/onci.22009
  • Weiner LM, Belldegrun AS, Crawford J, Tolcher AW, Lockbaum P, Arends RH, et al. Dose and schedule study of panitumumab monotherapy in patients with advanced solid malignancies. Clin Cancer Res 2008; 14:502 - 8; http://dx.doi.org/10.1158/1078-0432.CCR-07-1509; PMID: 18223225
  • Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov 2004; 3:391 - 400; http://dx.doi.org/10.1038/nrd1381; PMID: 15136787
  • Nimmerjahn F, Ravetch JV. Fcgamma receptors: old friends and new family members. Immunity 2006; 24:19 - 28; http://dx.doi.org/10.1016/j.immuni.2005.11.010; PMID: 16413920
  • Hubert P, Amigorena S. Antibody-dependent cell cytotoxicity in monoclonal antibody-mediated tumor immunotherapy. Oncoimmunology 2012; 1:103 - 5; http://dx.doi.org/10.4161/onci.1.1.17963; PMID: 22720225
  • Winiarska M, Glodkowska-Mrowka E, Bil J, Golab J. Molecular mechanisms of the antitumor effects of anti-CD20 antibodies. Front Biosci 2011; 16:277 - 306; http://dx.doi.org/10.2741/3688; PMID: 21196171
  • Dunkelberger JR, Song WC. Complement and its role in innate and adaptive immune responses. Cell Res 2010; 20:34 - 50; http://dx.doi.org/10.1038/cr.2009.139; PMID: 20010915
  • Zipfel PF, Skerka C. Complement regulators and inhibitory proteins. Nat Rev Immunol 2009; 9:729 - 40; PMID: 19730437
  • Coiffier B, Lepage E, Briere J, Herbrecht R, Tilly H, Bouabdallah R, et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med 2002; 346:235 - 42; http://dx.doi.org/10.1056/NEJMoa011795; PMID: 11807147
  • McLaughlin P, Grillo-López AJ, Link BK, Levy R, Czuczman MS, Williams ME, et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J Clin Oncol 1998; 16:2825 - 33; PMID: 9704735
  • Sorbye SW, Kilvaer T, Valkov A, Donnem T, Smeland E, Al-Shibli K, et al. High expression of CD20+ lymphocytes in soft tissue sarcomas is a positive prognostic indicator. Oncoimmunology 2012; 1:75 - 7; http://dx.doi.org/10.4161/onci.1.1.17825; PMID: 22720216
  • Seimetz D. Novel monoclonal antibodies for cancer treatment: the trifunctional antibody catumaxomab (removab). J Cancer 2011; 2:309 - 16; http://dx.doi.org/10.7150/jca.2.309; PMID: 21716847
  • Witzig TE, Gordon LI, Cabanillas F, Czuczman MS, Emmanouilides C, Joyce R, et al. Randomized controlled trial of yttrium-90-labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed B-cell non-Hodgkin’s lymphoma. J Clin Oncol 2002; 20:2453 - 63; http://dx.doi.org/10.1200/JCO.2002.11.076; PMID: 12011122
  • Kaminski MS, Estes J, Zasadny KR, Francis IR, Ross CW, Tuck M, et al. Radioimmunotherapy with iodine (131)I tositumomab for relapsed or refractory B-cell non-Hodgkin lymphoma: updated results and long-term follow-up of the University of Michigan experience. Blood 2000; 96:1259 - 66; PMID: 10942366
  • Melero I, Hervas-Stubbs S, Glennie M, Pardoll DM, Chen L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer 2007; 7:95 - 106; http://dx.doi.org/10.1038/nrc2051; PMID: 17251916
  • Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 2012; 11:215 - 33; http://dx.doi.org/10.1038/nrd3626; PMID: 22301798
  • Bercovici N, Trautmann A. Revisiting the role of T cells in tumor regression. Oncoimmunology 2012; 1:346 - 50; http://dx.doi.org/10.4161/onci.18800; PMID: 22737611
  • Waitz R, Fassò M, Allison JP. CTLA-4 blockade synergizes with cryoablation to mediate tumor rejection. Oncoimmunology 2012; 1:544 - 6; http://dx.doi.org/10.4161/onci.19442; PMID: 22754781
  • Younes A, Yasothan U, Kirkpatrick P. Brentuximab vedotin. Nat Rev Drug Discov 2012; 11:19 - 20; http://dx.doi.org/10.1038/nrd3629; PMID: 22212672
  • Senter PD, Sievers EL. The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat Biotechnol 2012; 30:631 - 7; http://dx.doi.org/10.1038/nbt.2289; PMID: 22781692
  • Coleman RE. Bone cancer in 2011: Prevention and treatment of bone metastases. Nat Rev Clin Oncol 2011; 9:76 - 8; http://dx.doi.org/10.1038/nrclinonc.2011.198; PMID: 22182971
  • Ohtsu A, Shah MA, Van Cutsem E, Rha SY, Sawaki A, Park SR, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled Phase III study. J Clin Oncol 2011; 29:3968 - 76; http://dx.doi.org/10.1200/JCO.2011.36.2236; PMID: 21844504
  • Ready NE, Dudek AZ, Pang HH, Hodgson LD, Graziano SL, Green MR, et al. Cisplatin, irinotecan, and bevacizumab for untreated extensive-stage small-cell lung cancer: CALGB 30306, a Phase II study. J Clin Oncol 2011; 29:4436 - 41; http://dx.doi.org/10.1200/JCO.2011.35.6923; PMID: 21969504
  • Brufsky AM, Hurvitz S, Perez E, Swamy R, Valero V, O’Neill V, et al. RIBBON-2: a randomized, double-blind, placebo-controlled, Phase III trial evaluating the efficacy and safety of bevacizumab in combination with chemotherapy for second-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol 2011; 29:4286 - 93; http://dx.doi.org/10.1200/JCO.2010.34.1255; PMID: 21990397
  • Konner JA, Grabon DM, Gerst SR, Iasonos A, Thaler H, Pezzulli SD, et al. Phase II study of intraperitoneal paclitaxel plus cisplatin and intravenous paclitaxel plus bevacizumab as adjuvant treatment of optimal stage II/III epithelial ovarian cancer. J Clin Oncol 2011; 29:4662 - 8; http://dx.doi.org/10.1200/JCO.2011.36.1352; PMID: 22067389
  • Kim KB, Sosman JA, Fruehauf JP, Linette GP, Markovic SN, McDermott DF, et al. BEAM: a randomized Phase II study evaluating the activity of bevacizumab in combination with carboplatin plus paclitaxel in patients with previously untreated advanced melanoma. J Clin Oncol 2012; 30:34 - 41; http://dx.doi.org/10.1200/JCO.2011.34.6270; PMID: 22124101
  • Lee NY, Zhang Q, Pfister DG, Kim J, Garden AS, Mechalakos J, et al. Addition of bevacizumab to standard chemoradiation for locoregionally advanced nasopharyngeal carcinoma (RTOG 0615): a phase 2 multi-institutional trial. Lancet Oncol 2012; 13:172 - 80; http://dx.doi.org/10.1016/S1470-2045(11)70303-5; PMID: 22178121
  • Burger RA, Brady MF, Bookman MA, Fleming GF, Monk BJ, Huang H, et al, Gynecologic Oncology Group. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med 2011; 365:2473 - 83; http://dx.doi.org/10.1056/NEJMoa1104390; PMID: 22204724
  • Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, et al, ICON7 Investigators. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med 2011; 365:2484 - 96; http://dx.doi.org/10.1056/NEJMoa1103799; PMID: 22204725
  • Meyerhardt JA, Li L, Sanoff HK, Carpenter W 4th, Schrag D. Effectiveness of bevacizumab with first-line combination chemotherapy for Medicare patients with stage IV colorectal cancer. J Clin Oncol 2012; 30:608 - 15; http://dx.doi.org/10.1200/JCO.2011.38.9650; PMID: 22253466
  • von Minckwitz G, Eidtmann H, Rezai M, Fasching PA, Tesch H, Eggemann H, et al, German Breast Group, Arbeitsgemeinschaft Gynäkologische Onkologie–Breast Study Groups. Neoadjuvant chemotherapy and bevacizumab for HER2-negative breast cancer. N Engl J Med 2012; 366:299 - 309; http://dx.doi.org/10.1056/NEJMoa1111065; PMID: 22276820
  • Bear HD, Tang G, Rastogi P, Geyer CE Jr., Robidoux A, Atkins JN, et al. Bevacizumab added to neoadjuvant chemotherapy for breast cancer. N Engl J Med 2012; 366:310 - 20; http://dx.doi.org/10.1056/NEJMoa1111097; PMID: 22276821
  • Pierga JY, Petit T, Delozier T, Ferrero JM, Campone M, Gligorov J, et al. Neoadjuvant bevacizumab, trastuzumab, and chemotherapy for primary inflammatory HER2-positive breast cancer (BEVERLY-2): an open-label, single-arm phase 2 study. Lancet Oncol 2012; 13:375 - 84; http://dx.doi.org/10.1016/S1470-2045(12)70049-9; PMID: 22377126
  • Uldrick TS, Wyvill KM, Kumar P, O’Mahony D, Bernstein W, Aleman K, et al. Phase II study of bevacizumab in patients with HIV-associated Kaposi’s sarcoma receiving antiretroviral therapy. J Clin Oncol 2012; 30:1476 - 83; http://dx.doi.org/10.1200/JCO.2011.39.6853; PMID: 22430271
  • Kelly WK, Halabi S, Carducci M, George D, Mahoney JF, Stadler WM, et al. Randomized, double-blind, placebo-controlled phase III trial comparing docetaxel and prednisone with or without bevacizumab in men with metastatic castration-resistant prostate cancer: CALGB 90401. J Clin Oncol 2012; 30:1534 - 40; http://dx.doi.org/10.1200/JCO.2011.39.4767; PMID: 22454414
  • Aghajanian C, Blank SV, Goff BA, Judson PL, Teneriello MG, Husain A, et al. OCEANS: a randomized, double-blind, placebo-controlled phase III trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent epithelial ovarian, primary peritoneal, or fallopian tube cancer. J Clin Oncol 2012; 30:2039 - 45; http://dx.doi.org/10.1200/JCO.2012.42.0505; PMID: 22529265
  • Kindler HL, Karrison TG, Gandara DR, Lu C, Krug LM, Stevenson JP, et al. Multicenter, double-blind, placebo-controlled, randomized phase II trial of gemcitabine/cisplatin plus bevacizumab or placebo in patients with malignant mesothelioma. J Clin Oncol 2012; 30:2509 - 15; http://dx.doi.org/10.1200/JCO.2011.41.5869; PMID: 22665541
  • Morrow PK, Wulf GM, Ensor J, Booser DJ, Moore JA, Flores PR, et al. Phase I/II study of trastuzumab in combination with everolimus (RAD001) in patients with HER2-overexpressing metastatic breast cancer who progressed on trastuzumab-based therapy. J Clin Oncol 2011; 29:3126 - 32; http://dx.doi.org/10.1200/JCO.2010.32.2321; PMID: 21730275
  • Perez EA, Romond EH, Suman VJ, Jeong JH, Davidson NE, Geyer CE Jr., et al. Four-year follow-up of trastuzumab plus adjuvant chemotherapy for operable human epidermal growth factor receptor 2-positive breast cancer: joint analysis of data from NCCTG N9831 and NSABP B-31. J Clin Oncol 2011; 29:3366 - 73; http://dx.doi.org/10.1200/JCO.2011.35.0868; PMID: 21768458
  • Untch M, Fasching PA, Konecny GE, Hasmüller S, Lebeau A, Kreienberg R, et al. Pathologic complete response after neoadjuvant chemotherapy plus trastuzumab predicts favorable survival in human epidermal growth factor receptor 2-overexpressing breast cancer: results from the TECHNO trial of the AGO and GBG study groups. J Clin Oncol 2011; 29:3351 - 7; http://dx.doi.org/10.1200/JCO.2010.31.4930; PMID: 21788566
  • Slamon D, Eiermann W, Robert N, Pienkowski T, Martin M, Press M, et al, Breast Cancer International Research Group. Adjuvant trastuzumab in HER2-positive breast cancer. N Engl J Med 2011; 365:1273 - 83; http://dx.doi.org/10.1056/NEJMoa0910383; PMID: 21991949
  • Perez EA, Suman VJ, Davidson NE, Gralow JR, Kaufman PA, Visscher DW, et al. Sequential versus concurrent trastuzumab in adjuvant chemotherapy for breast cancer. J Clin Oncol 2011; 29:4491 - 7; http://dx.doi.org/10.1200/JCO.2011.36.7045; PMID: 22042958
  • Gianni L, Pienkowski T, Im YH, Roman L, Tseng LM, Liu MC, et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol 2012; 13:25 - 32; http://dx.doi.org/10.1016/S1470-2045(11)70336-9; PMID: 22153890
  • Untch M, Loibl S, Bischoff J, Eidtmann H, Kaufmann M, Blohmer JU, et al, German Breast Group (GBG), Arbeitsgemeinschaft Gynäkologische Onkologie-Breast (AGO-B) Study Group. Lapatinib versus trastuzumab in combination with neoadjuvant anthracycline-taxane-based chemotherapy (GeparQuinto, GBG 44): a randomised phase 3 trial. Lancet Oncol 2012; 13:135 - 44; http://dx.doi.org/10.1016/S1470-2045(11)70397-7; PMID: 22257523
  • Baselga J, Bradbury I, Eidtmann H, Di Cosimo S, de Azambuja E, Aura C, et al, NeoALTTO Study Team. Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet 2012; 379:633 - 40; http://dx.doi.org/10.1016/S0140-6736(11)61847-3; PMID: 22257673
  • Ebb D, Meyers P, Grier H, Bernstein M, Gorlick R, Lipshultz SE, et al. Phase II trial of trastuzumab in combination with cytotoxic chemotherapy for treatment of metastatic osteosarcoma with human epidermal growth factor receptor 2 overexpression: a report from the children’s oncology group. J Clin Oncol 2012; 30:2545 - 51; http://dx.doi.org/10.1200/JCO.2011.37.4546; PMID: 22665540
  • Blackwell KL, Burstein HJ, Storniolo AM, Rugo HS, Sledge G, Aktan G, et al. Overall survival benefit with lapatinib in combination with trastuzumab for patients with human epidermal growth factor receptor 2-positive metastatic breast cancer: final results from the EGF104900 Study. J Clin Oncol 2012; 30:2585 - 92; http://dx.doi.org/10.1200/JCO.2011.35.6725; PMID: 22689807
  • Coiffier B, Osmanov EA, Hong X, Scheliga A, Mayer J, Offner F, et al, LYM-3001 study investigators. Bortezomib plus rituximab versus rituximab alone in patients with relapsed, rituximab-naive or rituximab-sensitive, follicular lymphoma: a randomised phase 3 trial. Lancet Oncol 2011; 12:773 - 84; http://dx.doi.org/10.1016/S1470-2045(11)70150-4; PMID: 21724462
  • Sehn LH, MacDonald D, Rubin S, Cantin G, Rubinger M, Lemieux B, et al. Bortezomib ADDED to R-CVP is safe and effective for previously untreated advanced-stage follicular lymphoma: a phase II study by the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 2011; 29:3396 - 401; http://dx.doi.org/10.1200/JCO.2010.33.6594; PMID: 21810681
  • Fowler N, Kahl BS, Lee P, Matous JV, Cashen AF, Jacobs SA, et al. Bortezomib, bendamustine, and rituximab in patients with relapsed or refractory follicular lymphoma: the phase II VERTICAL study. J Clin Oncol 2011; 29:3389 - 95; http://dx.doi.org/10.1200/JCO.2010.32.1844; PMID: 21810687
  • Fischer K, Cramer P, Busch R, Stilgenbauer S, Bahlo J, Schweighofer CD, et al. Bendamustine combined with rituximab in patients with relapsed and/or refractory chronic lymphocytic leukemia: a multicenter phase II trial of the German Chronic Lymphocytic Leukemia Study Group. J Clin Oncol 2011; 29:3559 - 66; http://dx.doi.org/10.1200/JCO.2010.33.8061; PMID: 21844497
  • Watanabe T, Tobinai K, Shibata T, Tsukasaki K, Morishima Y, Maseki N, et al. Phase II/III study of R-CHOP-21 versus R-CHOP-14 for untreated indolent B-cell non-Hodgkin’s lymphoma: JCOG 0203 trial. J Clin Oncol 2011; 29:3990 - 8; http://dx.doi.org/10.1200/JCO.2011.34.8508; PMID: 21931035
  • Pfreundschuh M, Kuhnt E, Trümper L, Osterborg A, Trneny M, Shepherd L, et al, MabThera International Trial (MInT) Group. CHOP-like chemotherapy with or without rituximab in young patients with good-prognosis diffuse large-B-cell lymphoma: 6-year results of an open-label randomised study of the MabThera International Trial (MInT) Group. Lancet Oncol 2011; 12:1013 - 22; http://dx.doi.org/10.1016/S1470-2045(11)70235-2; PMID: 21940214
  • Récher C, Coiffier B, Haioun C, Molina TJ, Fermé C, Casasnovas O, et al, Groupe d’Etude des Lymphomes de l’Adulte. Intensified chemotherapy with ACVBP plus rituximab versus standard CHOP plus rituximab for the treatment of diffuse large B-cell lymphoma (LNH03-2B): an open-label randomised phase 3 trial. Lancet 2011; 378:1858 - 67; http://dx.doi.org/10.1016/S0140-6736(11)61040-4; PMID: 22118442
  • Salit RB, Fowler DH, Wilson WH, Dean RM, Pavletic SZ, Dunleavy K, et al. Dose-adjusted EPOCH-rituximab combined with fludarabine provides an effective bridge to reduced-intensity allogeneic hematopoietic stem-cell transplantation in patients with lymphoid malignancies. J Clin Oncol 2012; 30:830 - 6; http://dx.doi.org/10.1200/JCO.2011.37.0296; PMID: 22312100
  • Wang M, Fayad L, Wagner-Bartak N, Zhang L, Hagemeister F, Neelapu SS, et al. Lenalidomide in combination with rituximab for patients with relapsed or refractory mantle-cell lymphoma: a phase 1/2 clinical trial. Lancet Oncol 2012; 13:716 - 23; http://dx.doi.org/10.1016/S1470-2045(12)70200-0; PMID: 22677155
  • Kluin-Nelemans HC, Hoster E, Hermine O, Walewski J, Trneny M, Geisler CH, et al. Treatment of older patients with mantle-cell lymphoma. N Engl J Med 2012; 367:520 - 31; http://dx.doi.org/10.1056/NEJMoa1200920; PMID: 22873532
  • 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
  • Govindan R, Bogart J, Stinchcombe T, Wang X, Hodgson L, Kratzke R, et al. Randomized phase II study of pemetrexed, carboplatin, and thoracic radiation with or without cetuximab in patients with locally advanced unresectable non-small-cell lung cancer: Cancer and Leukemia Group B trial 30407. J Clin Oncol 2011; 29:3120 - 5; http://dx.doi.org/10.1200/JCO.2010.33.4979; PMID: 21747084
  • Maubec E, Petrow P, Scheer-Senyarich I, Duvillard P, Lacroix L, Gelly J, et al. Phase II study of cetuximab as first-line single-drug therapy in patients with unresectable squamous cell carcinoma of the skin. J Clin Oncol 2011; 29:3419 - 26; http://dx.doi.org/10.1200/JCO.2010.34.1735; PMID: 21810686
  • Pirker R, Pereira JR, von Pawel J, Krzakowski M, Ramlau R, Park K, et al. EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: analysis of data from the phase 3 FLEX study. Lancet Oncol 2012; 13:33 - 42; http://dx.doi.org/10.1016/S1470-2045(11)70318-7; PMID: 22056021
  • Weickhardt AJ, Price TJ, Chong G, Gebski V, Pavlakis N, Johns TG, et al. Dual targeting of the epidermal growth factor receptor using the combination of cetuximab and erlotinib: preclinical evaluation and results of the phase II DUX study in chemotherapy-refractory, advanced colorectal cancer. J Clin Oncol 2012; 30:1505 - 12; http://dx.doi.org/10.1200/JCO.2011.38.6599; PMID: 22412142
  • Tveit KM, Guren T, Glimelius B, Pfeiffer P, Sorbye H, Pyrhonen S, et al. Phase III trial of cetuximab with continuous or intermittent fluorouracil, leucovorin, and oxaliplatin (Nordic FLOX) versus FLOX alone in first-line treatment of metastatic colorectal cancer: the NORDIC-VII study. J Clin Oncol 2012; 30:1755 - 62; http://dx.doi.org/10.1200/JCO.2011.38.0915; PMID: 22473155
  • Dewdney A, Cunningham D, Tabernero J, Capdevila J, Glimelius B, Cervantes A, et al. Multicenter randomized phase II clinical trial comparing neoadjuvant oxaliplatin, capecitabine, and preoperative radiotherapy with or without cetuximab followed by total mesorectal excision in patients with high-risk rectal cancer (EXPERT-C). J Clin Oncol 2012; 30:1620 - 7; http://dx.doi.org/10.1200/JCO.2011.39.6036; PMID: 22473163
  • Carey LA, Rugo HS, Marcom PK, Mayer EL, Esteva FJ, Ma CX, et al. TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol 2012; 30:2615 - 23; http://dx.doi.org/10.1200/JCO.2010.34.5579; PMID: 22665533
  • van den Eertwegh AJ, Versluis J, van den Berg HP, Santegoets SJ, van Moorselaar RJ, van der Sluis TM, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 2012; 13:509 - 17; http://dx.doi.org/10.1016/S1470-2045(12)70007-4; PMID: 22326922
  • Madan RA, Mohebtash M, Arlen PM, Vergati M, Rauckhorst M, Steinberg SM, et al. Ipilimumab and a poxviral vaccine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 2012; 13:501 - 8; http://dx.doi.org/10.1016/S1470-2045(12)70006-2; PMID: 22326924
  • Margolin K, Ernstoff MS, Hamid O, Lawrence D, McDermott D, Puzanov I, et al. Ipilimumab in patients with melanoma and brain metastases: an open-label, phase 2 trial. Lancet Oncol 2012; 13:459 - 65; http://dx.doi.org/10.1016/S1470-2045(12)70090-6; PMID: 22456429
  • Lynch TJ, Bondarenko I, Luft A, Serwatowski P, Barlesi F, Chacko R, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study. J Clin Oncol 2012; 30:2046 - 54; http://dx.doi.org/10.1200/JCO.2011.38.4032; PMID: 22547592
  • Elter T, Gercheva-Kyuchukova L, Pylylpenko H, Robak T, Jaksic B, Rekhtman G, et al. Fludarabine plus alemtuzumab versus fludarabine alone in patients with previously treated chronic lymphocytic leukaemia: a randomised phase 3 trial. Lancet Oncol 2011; 12:1204 - 13; http://dx.doi.org/10.1016/S1470-2045(11)70242-X; PMID: 21992852
  • Pettitt AR, Jackson R, Carruthers S, Dodd J, Dodd S, Oates M, et al. Alemtuzumab in combination with methylprednisolone is a highly effective induction regimen for patients with chronic lymphocytic leukemia and deletion of TP53: final results of the national cancer research institute CLL206 trial. J Clin Oncol 2012; 30:1647 - 55; http://dx.doi.org/10.1200/JCO.2011.35.9695; PMID: 22493413
  • Smith MR, Saad F, Coleman R, Shore N, Fizazi K, Tombal B, et al. Denosumab and bone-metastasis-free survival in men with castration-resistant prostate cancer: results of a phase 3, randomised, placebo-controlled trial. Lancet 2012; 379:39 - 46; http://dx.doi.org/10.1016/S0140-6736(11)61226-9; PMID: 22093187
  • Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366:2443 - 54; http://dx.doi.org/10.1056/NEJMoa1200690; PMID: 22658127
  • Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366:2455 - 65; http://dx.doi.org/10.1056/NEJMoa1200694; PMID: 22658128
  • Kantarjian H, Thomas D, Jorgensen J, Jabbour E, Kebriaei P, Rytting M, et al. Inotuzumab ozogamicin, an anti-CD22-calecheamicin conjugate, for refractory and relapsed acute lymphocytic leukaemia: a phase 2 study. Lancet Oncol 2012; 13:403 - 11; http://dx.doi.org/10.1016/S1470-2045(11)70386-2; PMID: 22357140
  • Younes A, Kim S, Romaguera J, Copeland A, Farial SdeC, Kwak LW, et al. Phase I multidose-escalation study of the anti-CD19 maytansinoid immunoconjugate SAR3419 administered by intravenous infusion every 3 weeks to patients with relapsed/refractory B-cell lymphoma. J Clin Oncol 2012; 30:2776 - 82; http://dx.doi.org/10.1200/JCO.2011.39.4403; PMID: 22753910
  • Juergens H, Daw NC, Geoerger B, Ferrari S, Villarroel M, Aerts I, et al. Preliminary efficacy of the anti-insulin-like growth factor type 1 receptor antibody figitumumab in patients with refractory Ewing sarcoma. J Clin Oncol 2011; 29:4534 - 40; http://dx.doi.org/10.1200/JCO.2010.33.0670; PMID: 22025154
  • Malempati S, Weigel B, Ingle AM, Ahern CH, Carroll JM, Roberts CT, et al. Phase I/II trial and pharmacokinetic study of cixutumumab in pediatric patients with refractory solid tumors and Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol 2012; 30:256 - 62; http://dx.doi.org/10.1200/JCO.2011.37.4355; PMID: 22184397
  • Baselga J, Cortés J, Kim SB, Im SA, Hegg R, Im YH, et al, CLEOPATRA Study Group. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med 2012; 366:109 - 19; http://dx.doi.org/10.1056/NEJMoa1113216; PMID: 22149875
  • Cortés J, Fumoleau P, Bianchi GV, Petrella TM, Gelmon K, Pivot X, et al. Pertuzumab monotherapy after trastuzumab-based treatment and subsequent reintroduction of trastuzumab: activity and tolerability in patients with advanced human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol 2012; 30:1594 - 600; http://dx.doi.org/10.1200/JCO.2011.37.4207; PMID: 22393084
  • Jakubowiak AJ, Benson DM, Bensinger W, Siegel DS, Zimmerman TM, Mohrbacher A, et al. Phase I trial of anti-CS1 monoclonal antibody elotuzumab in combination with bortezomib in the treatment of relapsed/refractory multiple myeloma. J Clin Oncol 2012; 30:1960 - 5; http://dx.doi.org/10.1200/JCO.2011.37.7069; PMID: 22291084
  • Lonial S, Vij R, Harousseau JL, Facon T, Moreau P, Mazumder A, et al. Elotuzumab in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma. J Clin Oncol 2012; 30:1953 - 9; http://dx.doi.org/10.1200/JCO.2011.37.2649; PMID: 22547589
  • Diaz LA Jr., Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 2012; 486:537 - 40; PMID: 22722843
  • Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 2012; 486:532 - 6; PMID: 22722830
  • Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 2012; 483:100 - 3; http://dx.doi.org/10.1038/nature10868; PMID: 22281684
  • Ran S, He J, Huang X, Soares M, Scothorn D, Thorpe PE. Antitumor effects of a monoclonal antibody that binds anionic phospholipids on the surface of tumor blood vessels in mice. Clin Cancer Res 2005; 11:1551 - 62; http://dx.doi.org/10.1158/1078-0432.CCR-04-1645; PMID: 15746060
  • 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, Nomenclature Committee on Cell Death 2009. 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
  • Vitale I, Galluzzi L, Castedo M, Kroemer G. Mitotic catastrophe: a mechanism for avoiding genomic instability. Nat Rev Mol Cell Biol 2011; 12:385 - 92; http://dx.doi.org/10.1038/nrm3115; PMID: 21527953
  • Ran S, Downes A, Thorpe PE. Increased exposure of anionic phospholipids on the surface of tumor blood vessels. Cancer Res 2002; 62:6132 - 40; PMID: 12414638
  • Ran S, Thorpe PE. Phosphatidylserine is a marker of tumor vasculature and a potential target for cancer imaging and therapy. Int J Radiat Oncol Biol Phys 2002; 54:1479 - 84; http://dx.doi.org/10.1016/S0360-3016(02)03928-7; PMID: 12459374
  • Gerber DE, Stopeck AT, Wong L, Rosen LS, Thorpe PE, Shan JS, et al. Phase I safety and pharmacokinetic study of bavituximab, a chimeric phosphatidylserine-targeting monoclonal antibody, in patients with advanced solid tumors. Clin Cancer Res 2011; 17:6888 - 96; http://dx.doi.org/10.1158/1078-0432.CCR-11-1074; PMID: 21989064
  • Gopal AK, Pagel JM, Fromm JR, Wilbur S, Press OW. 131I anti-CD45 radioimmunotherapy effectively targets and treats T-cell non-Hodgkin lymphoma. Blood 2009; 113:5905 - 10; http://dx.doi.org/10.1182/blood-2009-02-205476; PMID: 19332764
  • Matthews DC, Appelbaum FR, Eary JF, Fisher DR, Durack LD, Hui TE, et al. Phase I study of (131)I-anti-CD45 antibody plus cyclophosphamide and total body irradiation for advanced acute leukemia and myelodysplastic syndrome. Blood 1999; 94:1237 - 47; PMID: 10438711
  • Nagorsen D, Kufer P, Baeuerle PA, Bargou R. Blinatumomab: A historical perspective. Pharmacol Ther 2012; 136:334 - 42; http://dx.doi.org/10.1016/j.pharmthera.2012.07.013; PMID: 22940266
  • Nagorsen D, Baeuerle PA. Immunomodulatory therapy of cancer with T cell-engaging BiTE antibody blinatumomab. Exp Cell Res 2011; 317:1255 - 60; http://dx.doi.org/10.1016/j.yexcr.2011.03.010; PMID: 21419116
  • Topp MS, Kufer P, Gökbuget N, Goebeler M, Klinger M, Neumann S, et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol 2011; 29:2493 - 8; http://dx.doi.org/10.1200/JCO.2010.32.7270; PMID: 21576633
  • Topp MS, Gökbuget N, Zugmaier G, Degenhardt E, Goebeler ME, Klinger M, et al. Long-term follow-up of hematological relapse-free survival in a phase 2 study of blinatumomab in patients with minimal residual disease (MRD) of B-precursor acute lymphoblastic leukemia (ALL). Blood 2012; In press http://dx.doi.org/10.1182/blood-2012-07-441030; PMID: 23024237
  • Mueller BM, Romerdahl CA, Gillies SD, Reisfeld RA. Enhancement of antibody-dependent cytotoxicity with a chimeric anti-GD2 antibody. J Immunol 1990; 144:1382 - 6; PMID: 2303711
  • Cheung NK, Guo H, Hu J, Tassev DV, Cheung IY. Humanizing murine IgG3 anti-GD2 antibody m3F8 substantially improves antibody-dependent cell-mediated cytotoxicity while retaining targeting in vivo. Oncoimmunology 2012; 1:477 - 86; http://dx.doi.org/10.4161/onci.19864; PMID: 22754766
  • Saleh MN, Khazaeli MB, Wheeler RH, Allen L, Tilden AB, Grizzle W, et al. Phase I trial of the chimeric anti-GD2 monoclonal antibody ch14.18 in patients with malignant melanoma. Hum Antibodies Hybridomas 1992; 3:19 - 24; PMID: 1576319
  • Handgretinger R, Anderson K, Lang P, Dopfer R, Klingebiel T, Schrappe M, et al. A phase I study of human/mouse chimeric antiganglioside GD2 antibody ch14.18 in patients with neuroblastoma. Eur J Cancer 1995; 31A:261 - 7; http://dx.doi.org/10.1016/0959-8049(94)00413-Y; PMID: 7718335
  • Murray JL, Kleinerman ES, Jia SF, Rosenblum MG, Eton O, Buzaid A, et al. Phase Ia/Ib trial of anti-GD2 chimeric monoclonal antibody 14.18 (ch14.18) and recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) in metastatic melanoma. J Immunother Emphasis Tumor Immunol 1996; 19:206 - 17; http://dx.doi.org/10.1097/00002371-199605000-00005; PMID: 8811495
  • Ozkaynak MF, Sondel PM, Krailo MD, Gan J, Javorsky B, Reisfeld RA, et al. Phase I study of chimeric human/murine anti-ganglioside G(D2) monoclonal antibody (ch14.18) with granulocyte-macrophage colony-stimulating factor in children with neuroblastoma immediately after hematopoietic stem-cell transplantation: a Children’s Cancer Group Study. J Clin Oncol 2000; 18:4077 - 85; PMID: 11118469
  • Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, et al, Children’s Oncology Group. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med 2010; 363:1324 - 34; http://dx.doi.org/10.1056/NEJMoa0911123; PMID: 20879881
  • Simon T, Hero B, Faldum A, Handgretinger R, Schrappe M, Klingebiel T, et al. Long term outcome of high-risk neuroblastoma patients after immunotherapy with antibody ch14.18 or oral metronomic chemotherapy. BMC Cancer 2011; 11:21; http://dx.doi.org/10.1186/1471-2407-11-21; PMID: 21244693
  • Rosenfeld RG. Insulin-like growth factors and the basis of growth. N Engl J Med 2003; 349:2184 - 6; http://dx.doi.org/10.1056/NEJMp038156; PMID: 14657423
  • Atzori F, Tabernero J, Cervantes A, Prudkin L, Andreu J, Rodríguez-Braun E, et al. A phase I pharmacokinetic and pharmacodynamic study of dalotuzumab (MK-0646), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in patients with advanced solid tumors. Clin Cancer Res 2011; 17:6304 - 12; http://dx.doi.org/10.1158/1078-0432.CCR-10-3336; PMID: 21810918
  • Naing A, Kurzrock R, Burger A, Gupta S, Lei X, Busaidy N, et al. Phase I trial of cixutumumab combined with temsirolimus in patients with advanced cancer. Clin Cancer Res 2011; 17:6052 - 60; http://dx.doi.org/10.1158/1078-0432.CCR-10-2979; PMID: 21750201
  • Naing A, LoRusso P, Fu S, Hong DS, Anderson P, Benjamin RS, et al. Insulin growth factor-receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with refractory Ewing’s sarcoma family tumors. Clin Cancer Res 2012; 18:2625 - 31; http://dx.doi.org/10.1158/1078-0432.CCR-12-0061; PMID: 22465830
  • Daher M, Lacouture ME, Rathkopf D, Maki RG, Keohan ML, Gansukh B, et al. Case series of dermatologic events associated with the insulin-like growth factor receptor 1 inhibitor cixutumumab. J Clin Oncol 2011; 29:e638 - 40; http://dx.doi.org/10.1200/JCO.2010.34.5116; PMID: 21606420
  • Weickhardt A, Doebele R, Oton A, Lettieri J, Maxson D, Reynolds M, et al. A phase I/II study of erlotinib in combination with the anti-insulin-like growth factor-1 receptor monoclonal antibody IMC-A12 (cixutumumab) in patients with advanced non-small cell lung cancer. J Thorac Oncol 2012; 7:419 - 26; http://dx.doi.org/10.1097/JTO.0b013e31823c5b11; PMID: 22237261
  • Kindler HL, Richards DA, Garbo LE, Garon EB, Stephenson JJ Jr., Rocha-Lima CM, et al. A randomized, placebo-controlled phase 2 study of ganitumab (AMG 479) or conatumumab (AMG 655) in combination with gemcitabine in patients with metastatic pancreatic cancer. Ann Oncol 2012; 23:2834 - 42; http://dx.doi.org/10.1093/annonc/mds142; PMID: 22700995
  • Murakami H, Doi T, Yamamoto N, Watanabe J, Boku N, Fuse N, et al. Phase 1 study of ganitumab (AMG 479), a fully human monoclonal antibody against the insulin-like growth factor receptor type I (IGF1R), in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol 2012; 70:407 - 14; http://dx.doi.org/10.1007/s00280-012-1924-9; PMID: 22810805
  • Rosen LS, Puzanov I, Friberg G, Chan E, Hwang YC, Deng H, et al. Safety and pharmacokinetics of ganitumab (AMG 479) combined with sorafenib, panitumumab, erlotinib, or gemcitabine in patients with advanced solid tumors. Clin Cancer Res 2012; 18:3414 - 27; http://dx.doi.org/10.1158/1078-0432.CCR-11-3369; PMID: 22510349
  • Tap WD, Demetri G, Barnette P, Desai J, Kavan P, Tozer R, et al. Phase II study of ganitumab, a fully human anti-type-1 insulin-like growth factor receptor antibody, in patients with metastatic Ewing family tumors or desmoplastic small round cell tumors. J Clin Oncol 2012; 30:1849 - 56; http://dx.doi.org/10.1200/JCO.2011.37.2359; PMID: 22508822
  • Yee D. Insulin-like growth factor receptor inhibitors: baby or the bathwater?. J Natl Cancer Inst 2012; 104:975 - 81; http://dx.doi.org/10.1093/jnci/djs258; PMID: 22761272
  • Trial is halted for ganitumab in pancreatic cancer. Cancer Discov 2012; 2:OF4; http://dx.doi.org/10.1158/2159-8290.CD-NB2012-100; PMID: 23071056
  • Kline J, Gajewski TF. Clinical development of mAbs to block the PD1 pathway as an immunotherapy for cancer. Curr Opin Investig Drugs 2010; 11:1354 - 9; PMID: 21154117
  • Karwacz K, Arce F, Bricogne C, Kochan G, Escors D. PD-L1 co-stimulation, ligand-induced TCR down-modulation and anti-tumor immunotherapy. Oncoimmunology 2012; 1:86 - 8; http://dx.doi.org/10.4161/onci.1.1.17824; PMID: 22318430
  • Haymaker C, Wu R, Bernatchez C, Radvanyi L. PD-1 and BTLA and CD8(+) T-cell “exhaustion” in cancer: “Exercising” an alternative viewpoint. Oncoimmunology 2012; 1:735 - 8; http://dx.doi.org/10.4161/onci.20823; PMID: 22934265
  • 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; http://dx.doi.org/10.1158/0008-5472.CAN-07-0538; PMID: 17616683
  • 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; http://dx.doi.org/10.1182/blood-2007-07-100362; PMID: 17925489
  • Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004; 350:2129 - 39; http://dx.doi.org/10.1056/NEJMoa040938; PMID: 15118073
  • Goldberg RM. Cetuximab. Nat Rev Drug Discov 2005; Suppl S10 - 1; http://dx.doi.org/10.1038/nrd1728; PMID: 15962524
  • Saltz L, Easley C, Kirkpatrick P. Panitumumab. Nat Rev Drug Discov 2006; 5:987 - 8; http://dx.doi.org/10.1038/nrd2204; PMID: 17201026
  • Pollack BP. EGFR inhibitors, MHC expression and immune responses : Can EGFR inhibitors be used as immune response modifiers?. Oncoimmunology 2012; 1:71 - 4; http://dx.doi.org/10.4161/onci.1.1.18073; PMID: 22720215
  • Bebb G, Smith C, Rorke S, Boland W, Nicacio L, Sukhoo R, et al. Phase I clinical trial of the anti-EGFR monoclonal antibody nimotuzumab with concurrent external thoracic radiotherapy in Canadian patients diagnosed with stage IIb, III or IV non-small cell lung cancer unsuitable for radical therapy. Cancer Chemother Pharmacol 2011; 67:837 - 45; http://dx.doi.org/10.1007/s00280-010-1379-9; PMID: 20563810
  • Choi HJ, Sohn JH, Lee CG, Shim HS, Lee IJ, Yang WI, et al. A phase I study of nimotuzumab in combination with radiotherapy in stages IIB-IV non-small cell lung cancer unsuitable for radical therapy: Korean results. Lung Cancer 2011; 71:55 - 9; http://dx.doi.org/10.1016/j.lungcan.2010.04.010; PMID: 20451284
  • Hong J, Peng Y, Liao Y, Jiang W, Wei R, Huo L, et al. Nimotuzumab prolongs survival in patients with malignant gliomas: A phase I/II clinical study of concomitant radiochemotherapy with or without nimotuzumab. Exp Ther Med 2012; 4:151 - 7; PMID: 23060940
  • Kuenen B, Witteveen PO, Ruijter R, Giaccone G, Dontabhaktuni A, Fox F, et al. A phase I pharmacologic study of necitumumab (IMC-11F8), a fully human IgG1 monoclonal antibody directed against EGFR in patients with advanced solid malignancies. Clin Cancer Res 2010; 16:1915 - 23; http://dx.doi.org/10.1158/1078-0432.CCR-09-2425; PMID: 20197484
  • Li LF, Wang HQ, Liu XM, Zhang HL, Qiu LH, Qian ZZ, et al. [Nimotuzumab in combination with chemotherapy in patients with advanced non-small cell lung cancer]. Zhonghua Zhong Liu Za Zhi 2011; 33:626 - 8; PMID: 22325226
  • Ling Y, Chen J, Tao M, Chu X, Zhang X. A pilot study of nimotuzumab combined with cisplatin and 5-FU in patients with advanced esophageal squamous cell carcinoma. J Thorac Dis 2012; 4:58 - 62; PMID: 22295168
  • Qi DL, Wang HQ, Li Y, Huang CB, Wang QS, Xu L, et al. [Efficacy and adverse effets of nimotuzumab plus palitaxel liposome and carboplatin in the treatment for advanced non-small cell lung cancer]. Zhonghua Zhong Liu Za Zhi 2012; 34:152 - 5; PMID: 22780937
  • Ramos-Suzarte M, Lorenzo-Luaces P, Lazo NG, Perez ML, Soriano JL, Gonzalez CE, et al. Treatment of malignant, non-resectable, epithelial origin esophageal tumours with the humanized anti-epidermal growth factor antibody nimotuzumab combined with radiation therapy and chemotherapy. Cancer Biol Ther 2012; 13:600 - 5; http://dx.doi.org/10.4161/cbt.19849; PMID: 22555809
  • Strumberg D, Schultheis B, Scheulen ME, Hilger RA, Krauss J, Marschner N, et al. Phase II study of nimotuzumab, a humanized monoclonal anti-epidermal growth factor receptor (EGFR) antibody, in patients with locally advanced or metastatic pancreatic cancer. Invest New Drugs 2012; 30:1138 - 43; http://dx.doi.org/10.1007/s10637-010-9619-8; PMID: 21170759
  • Verduzco-Rodríguez L, Aguirre-González EH, Verduzco-Aguirre HC. Durable complete response induced by paclitaxel-nimotuzumab-methotrexate chemotherapy in a patient with metastatic head and neck squamous cell carcinoma. Hematol Oncol Stem Cell Ther 2011; 4:182 - 4; PMID: 22198189
  • Yang QY, Shen D, Sai K, Mu YG, Jiang XB, Zhang XH, et al. [Nimotuzumab in combination with chemotherapy for patients with malignant gliomas]. Zhonghua Zhong Liu Za Zhi 2011; 33:232 - 5; PMID: 21575527
  • You B, Brade A, Magalhaes JM, Siu LL, Oza A, Lovell S, et al. A dose-escalation phase I trial of nimotuzumab, an antibody against the epidermal growth factor receptor, in patients with advanced solid malignancies. Invest New Drugs 2011; 29:996 - 1003; http://dx.doi.org/10.1007/s10637-010-9444-0; PMID: 20454832
  • Zhao KL, Hu XC, Wu XH, Fu XL, Fan M, Jiang GL. A phase I dose escalation study of Nimotuzumab in combination with concurrent chemoradiation for patients with locally advanced squamous cell carcinoma of esophagus. Invest New Drugs 2012; 30:1585 - 90; http://dx.doi.org/10.1007/s10637-011-9735-0; PMID: 21901403
  • Zhao XY, Guo Y, Zhu YX, Wang Y, Zhu GP, Hu CS, et al. [Clinical analysis of nimotuzumab plus cisplatin and fluorouracil regimen as induction treatment in resectable head and neck squamous cell carcinoma]. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 2012; 47:536 - 9; PMID: 22932235
  • Krupitskaya Y, Wakelee HA. Ramucirumab, a fully human mAb to the transmembrane signaling tyrosine kinase VEGFR-2 for the potential treatment of cancer. Curr Opin Investig Drugs 2009; 10:597 - 605; PMID: 19513949
  • Hsu JY, Wakelee HA. Monoclonal antibodies targeting vascular endothelial growth factor: current status and future challenges in cancer therapy. BioDrugs 2009; 23:289 - 304; http://dx.doi.org/10.2165/11317600-000000000-00000; PMID: 19754219
  • Spratlin JL, Cohen RB, Eadens M, Gore L, Camidge DR, Diab S, et al. Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J Clin Oncol 2010; 28:780 - 7; http://dx.doi.org/10.1200/JCO.2009.23.7537; PMID: 20048182
  • Garon EB, Cao D, Alexandris E, John WJ, Yurasov S, Perol MA. A Randomized, Double-Blind, Phase III Study of Docetaxel and Ramucirumab Versus Docetaxel and Placebo in the Treatment of Stage IV Non-Small-Cell Lung Cancer After Disease Progression After 1 Previous Platinum-Based Therapy (REVEL): Treatment Rationale and Study Design. Clin Lung Cancer 2012; 13:505 - 9; http://dx.doi.org/10.1016/j.cllc.2012.06.007; PMID: 22853980
  • van Zaanen HC, Lokhorst HM, Aarden LA, Rensink HJ, Warnaar SO, van der Lelie J, et al. Chimaeric anti-interleukin 6 monoclonal antibodies in the treatment of advanced multiple myeloma: a phase I dose-escalating study. Br J Haematol 1998; 102:783 - 90; http://dx.doi.org/10.1046/j.1365-2141.1998.00835.x; PMID: 9722307
  • Fizazi K, De Bono JS, Flechon A, Heidenreich A, Voog E, Davis NB, et al. Randomised phase II study of siltuximab (CNTO 328), an anti-IL-6 monoclonal antibody, in combination with mitoxantrone/prednisone versus mitoxantrone/prednisone alone in metastatic castration-resistant prostate cancer. Eur J Cancer 2012; 48:85 - 93; http://dx.doi.org/10.1016/j.ejca.2011.10.014; PMID: 22129890
  • Hudes G, Tagawa ST, Whang YE, Qi M, Qin X, Puchalski TA, et al. A phase 1 study of a chimeric monoclonal antibody against interleukin-6, siltuximab, combined with docetaxel in patients with metastatic castration-resistant prostate cancer. Invest New Drugs 2012; http://dx.doi.org/10.1007/s10637-012-9857-z; PMID: 22828917
  • Karkera J, Steiner H, Li W, Skradski V, Moser PL, Riethdorf S, et al. The anti-interleukin-6 antibody siltuximab down-regulates genes implicated in tumorigenesis in prostate cancer patients from a phase I study. Prostate 2011; 71:1455 - 65; http://dx.doi.org/10.1002/pros.21362; PMID: 21321981
  • Oldenhuis CN, Stegehuis JH, Walenkamp AM, de Jong S, de Vries EG. Targeting TRAIL death receptors. Curr Opin Pharmacol 2008; 8:433 - 9; http://dx.doi.org/10.1016/j.coph.2008.06.011; PMID: 18625341
  • Kim K, Fisher MJ, Xu SQ, el-Deiry WS. Molecular determinants of response to TRAIL in killing of normal and cancer cells. Clin Cancer Res 2000; 6:335 - 46; PMID: 10690508
  • Yada A, Yazawa M, Ishida S, Yoshida H, Ichikawa K, Kurakata S, et al. A novel humanized anti-human death receptor 5 antibody CS-1008 induces apoptosis in tumor cells without toxicity in hepatocytes. Ann Oncol 2008; 19:1060 - 7; http://dx.doi.org/10.1093/annonc/mdn015; PMID: 18304965
  • Forero-Torres A, Shah J, Wood T, Posey J, Carlisle R, Copigneaux C, et al. Phase I trial of weekly tigatuzumab, an agonistic humanized monoclonal antibody targeting death receptor 5 (DR5). Cancer Biother Radiopharm 2010; 25:13 - 9; http://dx.doi.org/10.1089/cbr.2009.0673; PMID: 20187792
  • Gulley JL, Dahut WL. Future directions in tumor immunotherapy: CTLA4 blockade. Nat Clin Pract Oncol 2007; 4:136 - 7; http://dx.doi.org/10.1038/ncponc0749; PMID: 17327854
  • Pere H, Tanchot C, Bayry J, Terme M, Taieb J, Badoual C, et al. Comprehensive analysis of current approaches to inhibit regulatory T cells in cancer. Oncoimmunology 2012; 1:326 - 33; http://dx.doi.org/10.4161/onci.18852; PMID: 22737608
  • Tarhini AA, Cherian J, Moschos SJ, Tawbi HA, Shuai Y, Gooding WE, et al. Safety and efficacy of combination immunotherapy with interferon alfa-2b and tremelimumab in patients with stage IV melanoma. J Clin Oncol 2012; 30:322 - 8; http://dx.doi.org/10.1200/JCO.2011.37.5394; PMID: 22184371
  • McNeel DG, Smith HA, Eickhoff JC, Lang JM, Staab MJ, Wilding G, et al. Phase I trial of tremelimumab in combination with short-term androgen deprivation in patients with PSA-recurrent prostate cancer. Cancer Immunol Immunother 2012; 61:1137 - 47; http://dx.doi.org/10.1007/s00262-011-1193-1; PMID: 22210552
  • Abès R, Teillaud JL. Modulation of tumor immunity by therapeutic monoclonal antibodies. Cancer Metastasis Rev 2011; 30:111 - 24; http://dx.doi.org/10.1007/s10555-011-9282-3; PMID: 21267767