Skip to main content
Oncoimmunology logoLink to Oncoimmunology
. 2012 Jan 1;1(1):28–37. doi: 10.4161/onci.1.1.17938

Trial Watch

Monoclonal antibodies in cancer therapy

Lorenzo Galluzzi 1,2,3, Erika Vacchelli 1,2,3, Wolf Hervé Fridman 4,5,6,7, Jerome Galon 4,5,6,7, Catherine Sautès-Fridman 4,6,8, Eric Tartour 9,5,10, Jessica Zucman-Rossi 9,5,11, Laurence Zitvogel 2,12, Guido Kroemer 1,5,6,7,13,*
PMCID: PMC3376976  PMID: 22720209

Abstract

Since the advent of hybridoma technology, dating back to 1975, monoclonal antibodies have become an irreplaceable diagnostic and therapeutic tool for a wide array of human diseases. During the last 15 years, several monoclonal antibodies (mAbs) have been approved by FDA for cancer therapy. These mAbs are designed to (1) activate the immune system against tumor cells, (2) inhibit cancer cell-intrinsic signaling pathways, (3) bring toxins in the close proximity of cancer cells, or (4) interfere with the tumor-stroma interaction. More recently, major efforts have been made for the development of immunostimulatory mAbs that either enhance cancer-directed immune responses or limit tumor- (or therapy-) driven immunosuppression. Some of these antibodies, which are thought to facilitate tumor eradication by initiating or sustaining a tumor-specific immune response, have already entered clinical trials. In this Trial Watch, we will review and discuss the clinical progress of the most important mAbs that are have entered clinical trials after January 2008.

Keywords: bevacizumab, dalotuzumab, ipilimumab, nimotuzumab, ramucirumab, trastuzumab

Introduction

In 1975, Georges Köhler and César Milstein published in Nature the procedure to generate hybridomas, which are hybrid cell lines resulting from the fusion between a specific antibody-producing B cell (which normally cannot be maintained in culture) and a myeloma cell (which, as a cancer cell, is immortalized and hence can grow in culture, but does not synthesize specific antibody chains).1 In doing so, Köhler and Milstein provided a method to generate high amounts of antibodies with the same specificity, that is monoclonal, thereby de facto revolutionizing an incredible number of research procedures and clinical applications.2 In 1984, thanks to this breakthrough discovery, Köhler and Milstein shared the Nobel Prize for Medicine or Physiology with Niels Jerne, who made other contributions to immunology.3

Antibodies are composed of three functional units, two antigen-binding fragments (Fabs) and one constant fragment (Fc). Each Fab is made up by the association of one heavy and one light immunoglobulin chain, and at the level of the Fab three hypervariable complementarity-determining regions (CDRs) form the antigen-binding sites and confer antigen specificity. The Fc results from the association of two heavy chains, and links antibodies to immune effector functions.4-7 Based on the sequence of their heavy chains, antibodies can be subdivided into five classes (i.e., IgM, IgG, IgD, IgA and IgE), which are characterized by distinct functional profiles.8 The profound impact that monoclonal antibodies (mAbs) had on medical research, diagnostics, and clinical applications derives, for the most part, from (1) their ability to selectively bind two antigens of the same type with high affinity, and (2) from their elevated stability, in vitro and in vivo. Compared with their polyclonal counterparts, which are obtained from the serum of animals upon immunization, mAbs are advantageous in that they exhibit a much higher specificity and are less prone to contamination by pathogens.2

Thus, the monoclonal antibody technology has generated or considerably improved uncountable diagnostic applications, including (but not limited to) epitope-specific immunoblotting, immunofluorescence and immunohistochemistry. Moreover, mAbs have already been successfully employed in vivo (in animal models of disease or patients) to: (1) neutralize circulating factors, (2) activate immune effector mechanisms against disease-sustaining cell populations, (3) antagonize disease-specific molecules or molecular cascades, (4) crosslink plasma membrane receptors and hence activate therapeutic (diseased cell-autonomous or not) signaling pathways, (5) bring radionuclides, prodrugs, toxins or drug-filled liposomes in the proximity of target cells.4

Initially, mAbs were entirely murine, implying that they were relatively immunogenic and hence that their usefulness in humans was limited by the rapid elicitation of a neutralizing anti-mAb immune response. This issue was progressively solved with the ever increasing substitution of murine sequences with human ones, which eventually led to the development of fully humanized mAbs.9 Of note, even fully human mAbs do not entirely evade the control by the host immune system, as they are normally capable of eliciting an anti-idiotype response.10 Additional strategies that have been implemented to improve the pharmacological and pharmacodynamic properties of mAbs include (1) reduced or absent fucosylation and/or site specific mutations of the Fc, to enhance antibody-dependent cellular cytotoxicity (ADCC, see below),11 complement-dependent cytotoxicity (CDC, see below),12 and/or antibody-dependent cellular phagocytosis (ADCP, see below),13 or to increase serum half-life (by favoring the binding to the neonatal Fc receptor, which prevents IgG degradation),14 (2) addition of sialylated glycans, to increase anti-inflammatory properties,11 and (3) isotype chimerism, to boost ADCP15 and CDC.16

Irrespective of these considerations, antibodies of all types (murine, chimeric and human) have been approved by FDA and by other international regulatory agencies for the treatment of several pathologies, including (but not limited to) autoimmune diseases and cancer.2,4 Just to mention a few examples: muromonab—the first mAb ever approved for use in humans (for the therapy of transplant rejection)—is murine,17 infliximab—an anti-TNFα mAb that is currently employed against rheumatoid arthritis and Crohn's disease - is chimeric,18,19 and ofatumumab - an anti-CD20 mAb that is used for the therapy of chronic lymphocytic leukemia - is entirely human.20 Interestingly, the suffix of the international non-proprietary names for mAbs denotes the antibody format, as follows: -omab, mouse IgG2; -ximab, chimeric IgG1; -zumab, humanized IgG1; -umab, human antibody from phage display or transgenic mice technology; -axomab, trifunctional (bispecific) mouse-rat hybrid mAb; -cept, Fc fusion protein; -stim, Fc fusion peptide (Table 1). Most mAbs that are currently approved for use in humans (irrespective of their indication) or under clinical evaluation belong to the IgG isotype.2,4

Table 1. International suffixes for mAbs and mAb-related therapeutics.

Suffix Type of Molecule
-axomab
Bispecific mouse-rate hybrid mAb
-cept
FC fusion protein
-omab
Mouse IgG2
-stim
Fc fusion peptide
-umab
Human antibody from phage display or transgenic mice technology
-ximab
Chimeric IgG1
-zumab Humanized IgG1

Abbreviations: Fc, constant fragment.

There are at least six classes of mAbs that are relevant for cancer therapy: (1) mAbs that directly inhibit tumor cell-autonomous pro-survival cascades (e.g., cetuximab and panitumumab, which inhibit the epidermal growth factor receptor, EGFR, and are currently approved for the treatment of colorectal cancer);21 (2) mAbs that interfere with the tumor-stroma interaction, thereby indirectly inhibiting tumor growth (e.g., bevacizumab, which blocks the vascular endothelial growth factor, VEGF, and is currently employed in the for the therapy of colorectal, breast, renal and lung cancer);22-24 (3) mAbs that binds to antigens expressed on the surface of tumor cells and function by selectively engaging immune effector mechanisms such as ADCC,4,5 ADCP,25 and CDC6,7 (e.g., rituximab, a naked anti-CD20 in use for the therapy of lymphoma);26,27 (4) trifunctional (bispecific) mAbs, which can bind two different antigens while remaining capable of engaging immune effector functions (e.g., catumaxomab, an anti-CD3, anti-EpCAM chimeric mAb currently approved for the treatment of malignant ascites in patient with EpCAM+ neoplasms);28 (5) immunoconjugates (e.g., 90Y-ibritumomab tiuxetan and 131I-tositumomab, radionuclide-coupled anti-CD20 mAbs that are used for the therapy of lymphoma);29,30 and (6) immunostimulatory mAbs, i.e., mAbs that facilitate the development of a tumor-specific immune response by targeting the cancer cell/immune system crosstalk and the signaling pathways that this crosstalk elicits. An interesting sub-group encompassing these latter two categories is represented by immunoconjugates between putative tumor antigens and mAbs that target receptors expressed on the surface of dendritic cells (DCs), including CLEC9A, DC-SIGN, DEC205.31-33 By delivering tumor proteins (or protein epitopes) to DCs, these molecules stimulate antigen processing and cross-presentation, thus driving the establishment of a cancer-specific immune response involving CD4+ and CD8+ cells that—at least in preclinical models—often results in vaccination.34 This approach has also provided promising results in the field of infectiology.35,36

Table 2 summarizes the mAbs that are currently approved for use in cancer therapy. In this Trial Watch, we will discuss the clinical progress of mAbs that entered clinical trials for this indication after January 2008.

Table 2. Monoclonal antibodies currently approved for cancer therapy*.

mAb Target Approved Type Indication(s)
Alemtuzumab
CD52
2001
Hzed IgG1
Chronic lymphocytic leukemia
Bevacizumab
VEGF
2004
Hzed IgG1
Colorectal, breast, renal
and lung cancer
Catumaxomab
CD3 and EpCAM
2009
M-R hybrid
Malignant ascites in patients with EpCAM-positive cancer
Cetuximab
EGFR
2004
C IgG1
Colorectal cancer
Gemtuzumab
CD33
2000
Hzed IgG4
Acute myeloid leukemia
(coupled with calicheamicin)
Ibritumomab tiuxetan
CD20
2002
M IgG1
Non-Hodgkin lymphoma
(coupled with 90Y or 111In)
Ipilimumab
CTLA-4
2011
H IgG1
Melanoma
Panitumumab
EGFR
2006
H IgG2
Colorectal cancer
Ofatumumab
CD20
2009
H IgG1
Chronic lymphocytic leukemia
Rituximab
CD20
1997
C IgG1
Non-Hodgkin lymphoma
Tositumomab
CD20
2003
H IgG1
Non-Hodgkin lymphoma
(naked or coupled with 131I)
Trastuzumab ERBB2 1998 Hzed IgG1 Breast cancer

Abbreviations: C, chimeric; CTLA-4, cytotoxic T lymphocyte antigen 4; EGFR, epidermal growth factor receptor; EpCAM, epithelial cell adhesion molecule; H, human; Hzed, humanized; M, murine; R, rat; VEGF, vascular endothelial growth factor. * by FDA or European Medicines Agency (EMA) at the day of submission.

Monoclonal Antibodies Under Advanced (Phase III-IV) Clinical Evaluation

More than 75% of the ongoing clinical trials that test mAbs as anticancer agents aim at assessing the safety and efficacy of mAbs that have already been commercialized (Table 2), either as off-label medications (i.e., for diseases other than those for which they were initially approved) or combined with various chemo- and radiotherapeutic regimens (source www.clinicaltrials.gov). In spite of the fact that these mAbs are approved for cancer therapy in humans, only a relatively low percentage of these studies (around 12%) is in the late phases (III-IV) of evaluation. Moreover, bevacizumab-based (31) and trastuzumab-based (18) trials account alone for more than half of phase III and IV studies involving FDA-approved mAbs.

As few as 13 phase III-IV clinical studies based on experimental (not yet FDA approved) mAbs were being performed on August 2011 (when this manuscript has been prepared). These trials involve a total of 8 distinct agents and 7 cancer-specific molecular targets (Table 3).

Table 3. Monoclonal antibodies currently under evaluation for cancer therapy in phase III-IV trials*.

mAb Target Type Identifier Phase Indication(s)
Brentuximab vedotin**
CD30
MMAE-conjugated
C IgG1
NCT01100502
III
Hodgkin lymphoma
Ch14.18
GD2
C IgG1
NCT01041638
III
Neuroblastoma
Denosumab***
RANKL
H IgG2
NCT01077154
III
High risk early breast cancer receiving neoadjuvant or adjuvant therapy
Ganitumab
IGF1R
H IgG1
NCT01231347
III
Metastatic adenocarcinoma of the pancreas
Necitumumab
EGFR
H IgG1
NCT00981058
III
NSCLC
NCT00982111
III
NSCLC
Nimotuzumab
EGFR
Hzed IgG1
NCT01074021
III
Nasopharyngeal cancer
NCT00957086
III
HNC
NCT01249352
III
Locally advanced esophageal cancer
NCT01402180
III
Esophageal squamous cell carcinoma
Ramucirumab
VEGFR2
H IgG1
NCT00917384
III
Metastatic gastric or gastresophageal junction adenocarcinoma.
NCT01140347
III
Hepatocellular carcinoma after 1st line therapy with sorafenib
Siltuximab IL-6 C IgG1 NCT01266811 III Relapsed or refractory multiple myeloma

Abbreviations: C, chimeric; EGFR, epidermal growth factor receptor; H, human; HNC, head and neck cancer; Hzed, humanized; IGF1R, insulin-like growth factor 1 receptor; IL, interleukin; MMAE, monometil auristatin E; NSCLC, non-small cell lung cancer; RANKL, RANK ligand; VEGFR2, vascular endothelial growth factor receptor 2. *started after January, 1st 2008 and not completed or terminated at the day of submission; **currently reviewed by FDA and EMA; ***approved by both FDA and EMA for the therapy of bone loss during osteoporosis.

Brentuximab vedotin

Brentuximab vedotin (SGN-35) is a chimeric IgG1 mAb coupled to 3–5 units of the antimitotic compound monometil auristatin E (MMAE).37 The mAb moiety of brentuximab targets CD30 (TNFRSF8), a cell membrane protein of the tumor necrosis factor receptor (TNFR) family that is overexpressed by anaplastic large-cell and Hodgkin lymphoma.38 Contrarily to previous CD30-targeting approaches, brentuximab vedotin was found to induce durable objective responses and resulted in tumor regression in most patients with relapsed or refractory CD30-positive lymphomas.39 Today, brentuximab vedotin is being investigated in patients at high risk of residual Hodgkin lymphoma following stem cell transplant (Table 3, NCT01100502).

Ch14.18

Ch14.18 is a chimeric IgG1 mAb that targets disialoganglioside GD2, which often is present in high amounts at the surface of tumor cells of neuroendocrine origin.40 Characterized by an improved capacity to trigger ADCC (in particular by neutrophils)41 compared with other GD2-targeting mAbs,40 Ch14.18 entered phase I clinical trials for the treatment of melanoma and neuroblastoma in the early 1990s,42,43 with rather unsatisfactory results. Driven by preclinical data suggesting that granulocyte macrophage-colony stimulating factor (GM-CSF) exacerbates neuroblastoma cell killing by Ch14.18,41 several clinical trials were initiated later on to test Ch14.18 in combination with GM-CSF alone or GM-CSF + interleukin-2 (IL-2)44-46 Promising results have only been obtained when Ch14.18 was associated with GM-CSF, IL-1 and isotretinoin (a retinoid), leading to significantly improved outcome as compared with standard therapy in patients with high-risk neuroblastoma.46 As an alternative strategy, which never entered clinical tests, a Ch14.18-GM-CSF fusion protein was created, displaying comparable activity against neuroblastoma, in vitro, than Ch14.18 plus GM-CSF, and a good profile of ADCC and CDC induction.47 Now, Ch14.18 is being tested in combination with GM-CSF, IL-2, and isotretinoin after autologous stem cell transplant in neuroblastoma patients (Table 3, NCT01041638).

Denosumab

Denosumab (AMG 162) is a fully human IgG2 mAb currently approved by FDA for use in postmenopausal women with risk of osteoporosis. Denosumab specifically targets the receptor activator of NFκB ligand (RANKL), thus inhibiting the development and activity of osteoclasts, decreasing bone resorption and increasing bone density.48 As bone destruction is also an oncological problem (bones are a common site for metastases), it has been hypothesized that the anti-resorptive properties of denosumab might also be beneficial to cancer patients.49 Results from recent clinical trials suggest that denosumab can help prevent bone metastases in prostate cancer better than zoledronate, a bisphosphonate that had previously been shown to be superior to denosumab in the therapy of multiple myeloma.50 Today, denosumab is under evaluation for the prevention of bone metastasis in women with high risk early breast cancer receiving neoadjuvant or adjuvant therapy (Table 3, NCT01077154).

Ganitumab

Also known as AMG 479, ganitumab is a fully human IgG1 mAb directed against the insulin-like growth factor 1 receptor (IGF1R), a tyrosine kinase receptor that plays a critical role in oncogenic transformation.51 The IGF1R is highly overexpressed in most, if not all, malignant tissues, where it functions as an anti-apoptotic signal transducer.52 In vitro, ganitumab has been shown to virtually abolish ligand-induced IGF1R activation, thus limiting the viability of pancreatic cancer cells, in particular in conditions of serum deprivation. Ganitumab also inhibited tumor growth in vivo, in immunodeficient mice xenografted with human pancreatic cancer cells, suggesting that the antineoplastic effect of this mAb mostly rely on the inhibition of cell-autonomous signaling pathways.51 According to phase I clinical tests, there seem to be no major safety concerns for the use of ganitumab,53 which is now being tested in several phase I-II studies as well in a phase III trial (coupled with gemcitabine) for the therapy of metastatic adenocarcinomas of the pancreas (Table 3, NCT01231347).

Necitumumab

Necitumumab (IMC-11F8) is a fully human IgG1 mAb targeting the epidermal growth factor receptor (EGFR). A number of distinct tumors including colorectal and non small-cell lung cancer (NSCLC) exhibit activating mutations of the EGFR and several agents that inhibit the EGFR are approved as anticancer agents in the US and Europe. These include small cell-permeant chemicals that inhibit the tyrosine kinase activity of the receptor, like erlotinib and gefinitib,54-56 as well as the anti-EGFR mAbs cetuximab and panitumumab (Table 2), which block ligand-induced EGFR activation.57,58 Necitumumab shares this latter mechanism of action with cetuximab and panitumumab, and it has been shown to be well tolerated by patients and to achieve biologically relevant concentrations throughout the dosing period.59 Now, necitumumab is being evaluated in two distinct phase III trials for its efficiency against NSCLC, in combination with cisplatin (a DNA-damaging agent) and either gemcitabine or pemetrexed (an antimetabolite) (Table 3, NCT00981058 and NCT00982111, respectively).

Nimotuzumab

Similar to necitumumab, cetuximab and panitumumab, nimotuzumab is an anti-EGFR mAb. Developed in the early 2000s, nimotuzumab (a humanized IgG1 mAb also known as h-R3) soon demonstrated consistent in vitro and in vivo anticancer properties,60 boosting an intense wave of clinical test, often relying on the combination of nimotuzumab with radiotherapy.61-64 In parallel, nimotuzumab was conjugated to radionuclides including 188Os, 99mTc, 188Re and 177Lu, and some of the resulting immunoradioconjugates entered clinical tests as diagnostic or therapeutic tools for tumors of epidermal origin.65-68 Now, approximately 20 phase I-II clinical studies are investigating the efficacy of nimotuzumab for indications as diverse as glioma, head and neck, colorectal and lung cancer. Moreover, nimotuzumab is being evaluated in phase III trials (1) for the therapy of nasopharyngeal and head and neck cancer (HNC), in combination with not better specified chemotherapy and radiation (Table 2, NCT01074021, NCT00957086); (2) for the treatment of locally advanced esophageal cancer, in combination with 5-fluorouracil (a pyrimidine analog) and radiotherapy (Table 2, NCT01249352); and (3) in esophageal cancer patients who suffer with recurrence in regional lymph nodes after esophagectomy, combined with specified chemotherapy and radiation (Table 2, NCT01402180).

Ramucirumab

Ramucirumab (IMC-1121B) is a fully human IgG1 that specifically binds the VEGF receptor 2 (VEGFR2), thus antagonizing endogenous VEGF signaling.69 Similar to bevacizumab, which directly binds to and neutralizes circulating VEGF,70 ramucirumab was developed to inhibit perhaps the most important tumor-stroma interaction of all, angiogenesis.71 Ramucirumab was well tolerated in phase I clinical trials in patients with advanced cancers, and its most frequent mechanism-related dose-limiting toxicities were found to be hypertension and deep venous thrombosis.69,72 These studies also revealed promising rates of partial responses and stable disease, soliciting further clinical tests. Now, ramucirumab is under evaluation in multiple phase I-II trials as well as in two distinct phase II studies, one as a standalone intervention in patients with adenocarcinomas of the gastresophageal junction (Table 2, NCT00917384), and one as 2nd line treatment in patients with hepatocellular carcinoma after 1st line therapy with the chemical VEGFR2 inhibitor sorafenib (Table 2, NCT01140347).

Siltuximab

Siltuximab (CNTO-328) is a chimeric IgG1 mAb that binds to interleukin-6, a pleiotropic cytokine that is known to influence proliferation, apoptosis, and angiogenesis in cancer.73 However, contrarily to VEGF-targeting antibodies, siltuximab not only inhibits tumor growth by blocking the tumor-stroma interaction but also by interfering with tumor cell-autonomous signaling pathways, and notably with an MCL-1-mediated IL-6-driven antiapoptotic autocrine loop.74,75 Results from the first phase I clinical trial based on siltuximab were published as early as in 1998,73 yet more advanced studies lagged for another decade, with results from phase II studies being reported in the early 2010s,76-79 along with an ever more refined pre-clinical characterization of this mAb.80-84 Siltuximab is currently being tested in a phase III study in combination with the proteasomal inhibitor bortezomib and dexamethasone (a synthetic glucocorticoid) for the treatment of subjects with relapsed or refractory multiple myeloma (Table 2, NCT01266811).

Monoclonal Antibodies Under Early (phase I-II) Clinical Evaluation

There are around 200 open phase I-II clinical trials that test investigational (non FDA-approved) mAbs for oncological indications. Approximately one third of these studies is based on mAbs that are also being evaluated in phase III-IV trials (Table 3). As for the rest, there is a great heterogeneity of approaches, with some prominent trends that we will discuss here (source www.clinicaltrials.gov).

Cixutumumab (fully human IgG1) and dalotuzumab (humanized IgG1), two anti-IGF1R mAbs,85,86 are now being tested in 35 phase I-II clinical studies. By taking into account ganitumab (see above), the number of early clinical studies involving IGFR1-targeted therapies raises to more than 50, indicating that the expectations from this therapeutic approach are very high.

Bavituximab is a chimeric IgG1 specific for phosphatidylserine (PS), an anionic phospholipid that normally resides in the inner leaflet of the plasma membrane (from where it cannot be bound by mAbs).87 PS get exposed on the cell surface in some instances of cell death,88,89 cell activation and malignant transformation, and has been suggested to constitute a specific marker of tumor vasculature.90,91 In preclinical studies, bavituximab retarded the growth of different human tumors xenotransplanted in immunodeficient mice while eliciting no evident toxicity in the host,87 which supported its evaluation in the clinical setting. Now, the safety and efficiency of bavituximab, alone or in combination with conventional chemo-and radiotherapeutic regimens, are being investigated in 7 phase I-II trials, in a wide array of solid tumors (Table 4).

Table 4. Examples of monoclonal antibodies currently under evaluation for cancer therapy in phase I-II trials.

mAb
Target
Type
Trials*
Phase
Main indication(s)
AHN-12
CD45
90Y-conjugated
Hzed IgG1
1
I
Advanced leukemia.
Bavituximab
PS
C IgG1
7
I-II
Breast cancer; liver cancer; NSCLC; pancreatic cancer; prostate cancer.
BC8
CD45
90Y- or 131I-conjugated M IgG1
4
I-II
Acute lymphoblastic leukemia; AML; Hodgkin and non-Hodgkin lymphoma; myelodysplastic syndrome.
Blinatumomab
CD3 and CD19
na
2
II
B-precursor acute lymphoblastic leukemia.
Claudiximab
GC128
C IgG1
1
II
Advanced gastresophageal cancer
Conatumumab
TRAILR2
H IgG1
2
I-II
Colorectal cancer; lymphoma; NSCLC; pancreatic cancer; solid tumors.
Cixutumumab
IGF1R
H IgG1
26
I-II
Adrenocortical carcinoma; breast cancer; colorectal cancer; HNC; islet cell cancer; liver cancer; neuroendocrine cancer; NSCLC; pancreatic cancer; prostate cancer; sarcoma; SCLC; solid tumors.
c.T84.66
CEA
90Y-conjugated
C IgG1
2
I-II
Colorectal cancer; NSCLC.
Dalotuzumab
IGF1R
Hzed IgG1
9
I-II
Advanced solid tumors; breast cancer; colorectal cancer; NSCLC; pancreatic cancer; SCLC.
Ensituximab
NPC-1C
C IgG1
1
I
Colorectal cancer; pancreatic cancer.
Fresolimumab
TGFβ
H IgG4
3
I-II
Breast cancer; kidney cancer; melanoma; mesothelioma.
J591
PSMA
177Lu-conjugated
Hzed IgG1
2
II
Prostate cancer; solid tumors.
KB004
EphA
Hzed IgG1
1
I
Hematologic malignancies.
Lintuzumab
CD33
225Ac-conjugated or naked Hzed IgG1
2
I-II
Leukemia; myelodysplastic syndrome.
M5A
CEA
90Y-conjugated
Hzed IgG1
3
I-II
Colorectal cancer; solid tumors.
MDX-1106
PD1
Hzed IgG4
7
I-II
Melanoma; renal cell carcinoma; solid tumors.
MT110
CD3 and EpCAM
na
1
I
Advanced solid tumors
Olaratumab
PDGFRα
H IgG1
7
I-II
GIST; glioblastoma multiforme; NSCLC; prostate cancer; ovarian cancer; soft tissue sarcoma; solid tumors.
RAV12
RAAG12
C IgG1
1
II
Pancreatic cancer.
SAR650984
CD38
Hzed IgG1
1
I
CD38+ malignancies.
TF2
CEA and
IMP288
na
4
I-II
Colorectal cancer; SCLC.
Tigatuzumab
TRAILR2
Hzed IgG1
1
II
Triple negative breast cancer.
Tremelimumab
(Ticilimumab)
CTLA-4 H IgG2 4 I-II Advanced hepatocellular carcinoma; prostate cancer; recurrent melanoma.

Abbreviations: AML, acute myeloid leukemia; C, chimeric; CEA, carcinoembryonic antigen; CTLA-4, cytotoxic T lymphocyte antigen 4; EGFR, epidermal growth factor receptor; EpCAM, epithelial cell adhesion molecule; GIST, gastrointestinal stromal tumors; H, human; HNC, head and neck cancer; Hzed, humanized; IGF1R, insulin-like growth factor 1 receptor; M, murine; na, not applicable; NSCLC, non-small cell lung carcinoma; PD1, programmed death 1; PDGFR, platelet-derived growth factor receptor; PS, phosphatidylserine; PSMA, prostate-specific membrane antigen; SCLC, small cell lung carcinoma; TGFβ, transforming growth factor β; TRAILR2, tumor necrosis factor-related apoptosis-inducing ligand receptor 2. *total n° of trials started after January, 1st 2008 and not completed or terminated at the day of submission.

Olaratumab (IMC-3G3) is a fully human IgG1 antibody that specifically binds the α subunit of the platelet-derived growth factor receptor (PDGFRα), thus antagonizing endogenous PDGF signaling.92 PDGF receptors constitute promising targets for anticancer therapies as they are involved in the proliferation and survival of different tumors as well as in the regulation of the growth of tumor stroma and blood vessels.93 Blocking PDGFα with olaratumab on tumor (but not stromal) cells delayed tumor progression and reduced the size of skeletal metastases in preclinical models of pancreatic cancer.94 Now, olaratumab is being evaluated, either alone or combined with conventional chemotherapeutics, in 7 phase I-II trials against a variety of solid tumors (Table 4).

Similar to ipilimumab, a fully human IgG1 mAb currently approved for the therapy of melanoma (Table 2), tremelimumab (a human IgG2 also known as ticilimumab and CP-675,206) antagonizes the cytotoxic T lymphocyte antigen 4 (CTLA-4) on the surface of helper T cells, inhibiting the development of peripheral immune tolerance. Thus, tremelimumab belongs to the group of immunostimulatory mAbs, i.e., mAbs that do not directly target cancer cells but rather activate the immune system against them. Early clinical trials suggested that efficient doses of tremelimumab can be safely administered to humans, with major side-effects being autoimmune phenomena such as diarrhea, dermatitis, vitiligo, panhypopituitarism and hyperthyroidism.95 Several other phase I-II clinical studies have been conducted to evaluate the pharmacological profile of in tremelimumab in a wide array of solid tumors, all corroborating the encouraging results of preliminary tests.96-98 In spite of this, there appears to be no currently ongoing phase III-IV study based on tremelimumab, and only a few phase I-II studies are active (Table 4). Perhaps, this is not entirely unrelated to the success and commercialization of ipilimumab, which is now being tested in more than 30 clinical studies, all phases confounded.

Both conatumumab (human IgG1 also known as AMG 655) and tigatuzumab (humanized IgG1 also known as CS-1008) target the tumor necrosis factor-related apoptosis-inducing ligand receptor 2 (TRAILR2), a member of the death receptor protein family also known as death receptor 5 (DR5).99 Conatumumab and tigatuzumab operate as agonists, de facto activating TRAILR2 signaling and inducing the apoptotic demise of TRAILR2-expressing cancer (but not normal) cells.100,101 The exact mechanisms underlying the cancer-selective toxicity of TRAILR agonists remain to be fully elucidated, but may be related to the differential expression of TRAILRs and downstream signaling proteins in transformed vs. normal cells.102 Irrespective of these considerations, both conatumumab and tigatuzumab exhibited promising safety and efficacy profiles in preclinical tests and in initial phase I-II studies,100,103-105 which are now being extended to several oncological indications (Table 4).

MDX-1106 (ONO-4538) is a fully human IgG4 that specifically targets programmed death 1 (PD1), a transmembrane receptor that mediates immunosuppressive functions in activated T cells.106 Preclinical models suggested that the interruption of PD1 signaling leads to improved antitumor T cell responses and disease control,107 and stimulated a first wave of clinical tests, yielding promising results.108 MDX-1106 is now being investigated in 7 phase I-II studies, either alone or combined with vaccination strategies, for the therapy of solid tumors including melanoma and renal cell carcinoma (Table 4).

There are 14 distinct phase I-II trials currently assessing the safety and efficacy of bifunctional mAbs, including (but not limited to) the so-called BiTEs (bispecific T-cell engagers). BiTEs, such as the FDA-approved molecule catumaxomab (Table 2) and the investigational drugs blinatumomab and MT110 (Table 4), always target CD3 (a T lymphocyte transmembrane protein) plus one tumor-specific antigen (e.g., EpCAM in the case of catumaxomab and MT110, CD19 in the case of blinatumomab) and act as immunostimulatory agents by taking T cells in the close proximity of tumor cells.28,109,110 Another interesting approach that is being tested in phase I-II trials is known as pre-targeted radioimmunotherapy and is exemplified by TF2, a bispecific mAb that simultaneously targets the tumor-associated carcinoembryonic antigen (CEA) and a heterologous hapten peptide (IMP-288).111 In the context of preclinical studies that yielded encouraging results, TF2 was pre-administered to mice and allowed to localize to CEA-expressing tumors, followed by the injection of 177Lu-coupled IMP-288.112 Similar approaches are now being evaluated in patients affected by CEA-expressing malignancies, including colorectal and small cell lung cancer (Table 4).

Several distinct immunoconjugates, most of which carry radioactive isotopes, are the object of phase I-II studies (Table 4). These include (but are not limited to) CD45-targeting molecules (e.g., AHN-12 and BC8),113-115 which are being studied for the therapy of CD45-expressing hematopoietic cancers; immunoconjugates that specifically bind to CEA (e.g., c.T84.66 and M5A),116,117 which are being evaluated as a treatment for colorectal cancer and other solid tumors; and molecules that target the cell surface myelomonocytic differentiation antigen CD33 (e.g., lantuzumab, which—coupled to 225Ac—is under investigation in the context of hematopoietic malignancies).118 Of note, naked lantuzumab has previously been tested in phase III clinical trials, but failed to yield statistically significant improvements in response rates and patient survival.119,120 Another CD33-targeting immunoconjugate, gemtuzumab calicheamicin, is approved since 2000 for the treatment of acute myeloid leukemia (Table 2), possibly explaining why the interest in lantuzumab appears to be decreasing.

Some further examples of mAbs that are currently being investigated in phase I-II clinical trials for oncological indications are reported in Table 4.

Concluding Remarks

At the end of the 19th century, the immunologist Paul Ehrlich (Strehlen 1854; Bad Homburg 1915) hypothesized that several pathologies would be cured if “magic bullets” were available for selectively targeting the cause of disease. Thanks to the invention of mAbs,1 the “magic bullet” concept has evolved until becoming an ever more refined clinical reality. It can be anticipated that (at least) three major approaches will drive the development of the next generation of mAb-based therapies. First, novel cancer-specific targets will be identified, leading to the creation of mAbs with new specificities. Second, mAbs with known specificities will be engineered to improve their biological functions. Third, new mAbs that do not directly target cancer cells (including mAbs that interfere with the tumor-stroma interaction as well as immunostimulatory mAbs) will be discovered, along with the increasing knowledge that is being generated on the host-tumor crosstalk.

As discussed above, mAbs can function as anticancer agents by modulating tumor cell-intrinsic signaling cascades, by interfering with the tumor-stroma interaction, or by acutely activating the innate immune system (via ADCC, ADCP and CDC) against tumor cells. Intriguingly enough, emerging evidence indicates that—at least in some instances—mAbs exert therapeutic effects by triggering or enhancing a T cell-based response, aside from any effector mechanisms from innate immunity.121-123 In preclinical models, such an adaptive immune response appears to be required for the therapeutic effect of mAbs.124,125 The cellular and molecular circuitries underlying the induction of adaptive immunity by mAbs remain poorly understood.126 As a possibility, mAbs may enhance antigen uptake or cross-presentation by DCs,127,128 or facilitate immunogenic cell death.129 The precise elucidation of these aspects is eagerly awaited, as it could have profound implications for the development of novel mAbs.

Two additional elements that will have to be taken into consideration for the development of novel mAb-based therapies relate to the Fc receptor, and in particular to the facts that (1) some cancer cell types express functional Fc receptors, which can trigger unconventional (and hence presumably unwanted) signaling pathways,130-132 and that (2) the genes coding for human Fc receptors are very heterogeneous (there are 3 Fcγ receptor classes, specifying at least 12 receptor isoforms) and all of them present multiple polymorphisms that affect IgG binding and/or signal transduction efficiency.133 These polymorphisms have been shown to influence the patient response to FDA-approved mAbs that elicit immune effector functions, such as cetuximab and rituximab,134-136 calling for strategies to circumvent this issue. As mentioned above, approaches whereby the Fc fragment is subjected to specific mutations or to changes in its glycosylation status (i.e., reduced or absent fucosylation)137 are underway for resolving this problem.11 Future will tell which one among all these strategies will eventually lead to the generation of the next wave of FDA-approved mAbs.

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.

Glossary

Abbreviations:

ADCC

antibody-dependent cellular cytotoxicity

ADCP

antibody-dependent cellular phagocytosis

BiTEs

bispecific T-cell engagers

CDC

complement-dependent cytotoxicity

CDR

complementarity-determining region

CEA

carcinoembryonic antigen

DCs

dendritic cells

DR5

death receptor 5

EGFR

epidermal growth factor receptor

Fab

antigen-binding fragment

Fc

constant fragment

GM-CSF

granulocyte macrophage-colony stimulating factor

mAb

monoclonal antibody

MMAE

monometil auristatin E

NSCLC

non small-cell lung cancer

PDGFRα

plateled-derived growth factor receptor α

RANKL

receptor activator of NF-κB ligand

TNFR

tumor necrosis factor receptor

TRAILR2

tumor necrosis factor-related apoptosis-inducing ligand receptor 2

VEGFR

vascular endothelial growth factor receptor

Footnotes

References

  • 1.Köhler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256:495–7. doi: 10.1038/256495a0. [DOI] [PubMed] [Google Scholar]
  • 2.Alkan SS. Monoclonal antibodies: the story of a discovery that revolutionized science and medicine. Nat Rev Immunol. 2004;4:153–6. doi: 10.1038/nri1265. [DOI] [PubMed] [Google Scholar]
  • 3.Jerne NK. The generative grammar of the immune system. Science. 1985;229:1057–9. doi: 10.1126/science.4035345. [DOI] [PubMed] [Google Scholar]
  • 4.Weiner LM, Surana R, Wang S. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat Rev Immunol. 2010;10:317–27. doi: 10.1038/nri2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Nimmerjahn F, Ravetch JV. Fcgamma receptors: old friends and new family members. Immunity. 2006;24:19–28. doi: 10.1016/j.immuni.2005.11.010. [DOI] [PubMed] [Google Scholar]
  • 6.Dunkelberger JR, Song WC. Complement and its role in innate and adaptive immune responses. Cell Res. 2010;20:34–50. doi: 10.1038/cr.2009.139. [DOI] [PubMed] [Google Scholar]
  • 7.Zipfel PF, Skerka C. Complement regulators and inhibitory proteins. Nat Rev Immunol. 2009;9:729–40. doi: 10.1038/nri2620. [DOI] [PubMed] [Google Scholar]
  • 8.Metzger H. Molecular versatility of antibodies. Immunol Rev. 2002;185:186–205. doi: 10.1034/j.1600-065X.2002.18516.x. [DOI] [PubMed] [Google Scholar]
  • 9.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. doi: 10.1126/science.8235658. [DOI] [PubMed] [Google Scholar]
  • 10.Stephens S, Emtage S, Vetterlein O, Chaplin L, Bebbington C, Nesbitt A, et al. Comprehensive pharmacokinetics of a humanized antibody and analysis of residual anti-idiotypic responses. Immunology. 1995;85:668–74. [PMC free article] [PubMed] [Google Scholar]
  • 11.Jefferis R. Glycosylation as a strategy to improve antibody-based therapeutics. Nat Rev Drug Discov. 2009;8:226–34. doi: 10.1038/nrd2804. [DOI] [PubMed] [Google Scholar]
  • 12.Moore GL, Chen H, Karki S, Lazar GA. Engineered Fc variant antibodies with enhanced ability to recruit complement and mediate effector functions. MAbs. 2010;2:181–9. doi: 10.4161/mabs.2.2.11158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Horton HM, Bernett MJ, Peipp M, Pong E, Karki S, Chu SY, et al. Fc-engineered anti-CD40 antibody enhances multiple effector functions and exhibits potent in vitro and in vivo antitumor activity against hematologic malignancies. Blood. 2010;116:3004–12. doi: 10.1182/blood-2010-01-265280. [DOI] [PubMed] [Google Scholar]
  • 14.Roopenian DC, Akilesh S. FcRn: the neonatal Fc receptor comes of age. Nat Rev Immunol. 2007;7:715–25. doi: 10.1038/nri2155. [DOI] [PubMed] [Google Scholar]
  • 15.Nagashima H, Ootsubo M, Fukazawa M, Motoi S, Konakahara S, Masuho Y. Enhanced antibody-dependent cellular phagocytosis by chimeric monoclonal antibodies with tandemly repeated Fc domains. J Biosci Bioeng. 2011;111:391–6. doi: 10.1016/j.jbiosc.2010.12.007. [DOI] [PubMed] [Google Scholar]
  • 16.Natsume A, In M, Takamura H, Nakagawa T, Shimizu Y, Kitajima K, et al. Engineered antibodies of IgG1/IgG3 mixed isotype with enhanced cytotoxic activities. Cancer Res. 2008;68:3863–72. doi: 10.1158/0008-5472.CAN-07-6297. [DOI] [PubMed] [Google Scholar]
  • 17.A randomized clinical trial of OKT3 monoclonal antibody for acute rejection of cadaveric renal transplants. Ortho Multicenter Transplant Study Group. N Engl J Med. 1985;313:337–42. doi: 10.1056/NEJM198508083130601. [DOI] [PubMed] [Google Scholar]
  • 18.Lipsky PE, van der Heijde DM, St Clair EW, Furst DE, Breedveld FC, Kalden JR, et al. Infliximab and methotrexate in the treatment of rheumatoid arthritis. Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy Study Group. N Engl J Med. 2000;343:1594–602. doi: 10.1056/NEJM200011303432202. [DOI] [PubMed] [Google Scholar]
  • 19.Present DH, Rutgeerts P, Targan S, Hanauer SB, Mayer L, van Hogezand RA, et al. Infliximab for the treatment of fistulas in patients with Crohn's disease. N Engl J Med. 1999;340:1398–405. doi: 10.1056/NEJM199905063401804. [DOI] [PubMed] [Google Scholar]
  • 20.Keating MJ, Dritselis A, Yasothan U, Kirkpatrick P. Ofatumumab. Nat Rev Drug Discov. 2010;9:101–2. doi: 10.1038/nrd3100. [DOI] [PubMed] [Google Scholar]
  • 21.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. doi: 10.1158/1078-0432.CCR-07-1509. [DOI] [PubMed] [Google Scholar]
  • 22.Hurvitz SA, Allen HJ, Moroose RL, Chan D, Hagenstad C, Applebaum SH, et al. A phase II trial of docetaxel with bevacizumab as first-line therapy for HER2-negative metastatic breast cancer (TORI B01) Clin Breast Cancer. 2010;10:307–12. doi: 10.3816/CBC.2010.n.040. [DOI] [PubMed] [Google Scholar]
  • 23.Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, et al. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med. 2007;357:2666–76. doi: 10.1056/NEJMoa072113. [DOI] [PubMed] [Google Scholar]
  • 24.Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med. 2006;355:2542–50. doi: 10.1056/NEJMoa061884. [DOI] [PubMed] [Google Scholar]
  • 25.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. doi: 10.2741/3688. [DOI] [PubMed] [Google Scholar]
  • 26.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. doi: 10.1056/NEJMoa011795. [DOI] [PubMed] [Google Scholar]
  • 27.McLaughlin P, Grillo-Lopez 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. doi: 10.1200/JCO.1998.16.8.2825. [DOI] [PubMed] [Google Scholar]
  • 28.Seimetz D. Novel monoclonal antibodies for cancer treatment: the trifunctional antibody catumaxomab (removab) J Cancer. 2011;2:309–16. doi: 10.7150/jca.2.309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.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. doi: 10.1200/JCO.2002.11.076. [DOI] [PubMed] [Google Scholar]
  • 30.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. [PubMed] [Google Scholar]
  • 31.Tacken PJ, de Vries IJ, Gijzen K, Joosten B, Wu D, Rother RP, et al. Effective induction of naive and recall T-cell responses by targeting antigen to human dendritic cells via a humanized anti-DC-SIGN antibody. Blood. 2005;106:1278–85. doi: 10.1182/blood-2005-01-0318. [DOI] [PubMed] [Google Scholar]
  • 32.Charalambous A, Oks M, Nchinda G, Yamazaki S, Steinman RM. Dendritic cell targeting of survivin protein in a xenogeneic form elicits strong CD4+ T cell immunity to mouse survivin. J Immunol. 2006;177:8410–21. doi: 10.4049/jimmunol.177.12.8410. [DOI] [PubMed] [Google Scholar]
  • 33.Sancho D, Mourao-Sa D, Joffre OP, Schulz O, Rogers NC, Pennington DJ, et al. Tumor therapy in mice via antigen targeting to a novel, DC-restricted C-type lectin. J Clin Invest. 2008;118:2098–110. doi: 10.1172/JCI34584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tacken PJ, de Vries IJ, Torensma R, Figdor CG. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat Rev Immunol. 2007;7:790–802. doi: 10.1038/nri2173. [DOI] [PubMed] [Google Scholar]
  • 35.Stylianou E, Pepponi I, van Dolleweerd CJ, Paul MJ, Ma JK, Reljic R. Exploring the vaccine potential of Dec-205 targeting in Mycobacterium tuberculosis infection in mice. Vaccine. 2011;29:2279–86. doi: 10.1016/j.vaccine.2011.01.030. [DOI] [PubMed] [Google Scholar]
  • 36.Do Y, Koh H, Park CG, Dudziak D, Seo P, Mehandru S, et al. Targeting of LcrV virulence protein from Yersinia pestis to dendritic cells protects mice against pneumonic plague. Eur J Immunol. 2010;40:2791–6. doi: 10.1002/eji.201040511. [DOI] [PubMed] [Google Scholar]
  • 37.Ansell SM. Brentuximab vedotin: delivering an antimitotic drug to activated lymphoma cells. Expert Opin Investig Drugs. 2011;20:99–105. doi: 10.1517/13543784.2011.542147. [DOI] [PubMed] [Google Scholar]
  • 38.Deutsch YE, Tadmor T, Podack ER, Rosenblatt JD. CD30: an important new target in hematologic malignancies. Leuk Lymphoma. 2011;52:1641–54. doi: 10.3109/10428194.2011.574761. [DOI] [PubMed] [Google Scholar]
  • 39.Younes A, Bartlett NL, Leonard JP, Kennedy DA, Lynch CM, Sievers EL, et al. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N Engl J Med. 2010;363:1812–21. doi: 10.1056/NEJMoa1002965. [DOI] [PubMed] [Google Scholar]
  • 40.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. [PubMed] [Google Scholar]
  • 41.Barker E, Reisfeld RA. A mechanism for neutrophil-mediated lysis of human neuroblastoma cells. Cancer Res. 1993;53:362–7. [PubMed] [Google Scholar]
  • 42.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. [PubMed] [Google Scholar]
  • 43.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;31:261–7. doi: 10.1016/0959-8049(94)00413-Y. [DOI] [PubMed] [Google Scholar]
  • 44.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. doi: 10.1097/00002371-199605000-00005. [DOI] [PubMed] [Google Scholar]
  • 45.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. doi: 10.1200/JCO.2000.18.24.4077. [DOI] [PubMed] [Google Scholar]
  • 46.Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010;363:1324–34. doi: 10.1056/NEJMoa0911123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Batova A, Kamps A, Gillies SD, Reisfeld RA, Yu AL. The Ch14.18-GM-CSF fusion protein is effective at mediating antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity in vitro. Clin Cancer Res. 1999;5:4259–63. [PubMed] [Google Scholar]
  • 48.Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361:756–65. doi: 10.1056/NEJMoa0809493. [DOI] [PubMed] [Google Scholar]
  • 49.Vallet S, Smith MR, Raje N. Novel bone-targeted strategies in oncology. Clin Cancer Res. 2010;16:4084–93. doi: 10.1158/1078-0432.CCR-10-0600. [DOI] [PubMed] [Google Scholar]
  • 50.Weintraub B. Trials define anti-tumor effects of anti-resorptive agents: denosumab ahead of zoledronate 2 to 1. BioDrugs. 2011;25:135–8. doi: 10.2165/11590730-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 51.Beltran PJ, Mitchell P, Chung YA, Cajulis E, Lu J, Belmontes B, et al. AMG 479, a fully human anti-insulin-like growth factor receptor type I monoclonal antibody, inhibits the growth and survival of pancreatic carcinoma cells. Mol Cancer Ther. 2009;8:1095–105. doi: 10.1158/1535-7163.MCT-08-1171. [DOI] [PubMed] [Google Scholar]
  • 52.Rosenfeld RG. Insulin-like growth factors and the basis of growth. N Engl J Med. 2003;349:2184–6. doi: 10.1056/NEJMp038156. [DOI] [PubMed] [Google Scholar]
  • 53.Tolcher AW, Sarantopoulos J, Patnaik A, Papadopoulos K, Lin CC, Rodon J, et al. Phase I, pharmacokinetic, and pharmacodynamic study of AMG 479, a fully human monoclonal antibody to insulin-like growth factor receptor 1. J Clin Oncol. 2009;27:5800–7. doi: 10.1200/JCO.2009.23.6745. [DOI] [PubMed] [Google Scholar]
  • 54.de La Motte Rouge T, Galluzzi L, Olaussen KA, Zermati Y, Tasdemir E, Robert T, et al. A novel epidermal growth factor receptor inhibitor promotes apoptosis in non-small cell lung cancer cells resistant to erlotinib. Cancer Res. 2007;67:6253–62. doi: 10.1158/0008-5472.CAN-07-0538. [DOI] [PubMed] [Google Scholar]
  • 55.Boehrer S, Ades L, Braun T, Galluzzi L, Grosjean J, Fabre C, et al. Erlotinib exhibits antineoplastic off-target effects in AML and MDS: a preclinical study. Blood. 2008;111:2170–80. doi: 10.1182/blood-2007-07-100362. [DOI] [PubMed] [Google Scholar]
  • 56.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. doi: 10.1056/NEJMoa040938. [DOI] [PubMed] [Google Scholar]
  • 57.Goldberg RM. Cetuximab. Nat Rev Drug Discov. 2005;(Suppl):S10–1. doi: 10.1038/nrd1728. [DOI] [PubMed] [Google Scholar]
  • 58.Saltz L, Easley C, Kirkpatrick P. Panitumumab. Nat Rev Drug Discov. 2006;5:987–8. doi: 10.1038/nrd2204. [DOI] [PubMed] [Google Scholar]
  • 59.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. doi: 10.1158/1078-0432.CCR-09-2425. [DOI] [PubMed] [Google Scholar]
  • 60.Crombet-Ramos T, Rak J, Perez R, Viloria-Petit A. Antiproliferative, antiangiogenic and proapoptotic activity of h-R3: A humanized anti-EGFR antibody. Int J Cancer. 2002;101:567–75. doi: 10.1002/ijc.10647. [DOI] [PubMed] [Google Scholar]
  • 61.Crombet T, Torres L, Neninger E, Catala M, Solano ME, Perera A, et al. Pharmacological evaluation of humanized anti-epidermal growth factor receptor, monoclonal antibody h-R3, in patients with advanced epithelial-derived cancer. J Immunother. 2003;26:139–48. doi: 10.1097/00002371-200303000-00006. [DOI] [PubMed] [Google Scholar]
  • 62.Crombet T, Osorio M, Cruz T, Roca C, del Castillo R, Mon R, et al. Use of the humanized anti-epidermal growth factor receptor monoclonal antibody h-R3 in combination with radiotherapy in the treatment of locally advanced head and neck cancer patients. J Clin Oncol. 2004;22:1646–54. doi: 10.1200/JCO.2004.03.089. [DOI] [PubMed] [Google Scholar]
  • 63.Ramos TC, Figueredo J, Catala M, Gonzalez S, Selva JC, Cruz TM, et al. Treatment of high-grade glioma patients with the humanized anti-epidermal growth factor receptor (EGFR) antibody h-R3: report from a phase I/II trial. Cancer Biol Ther. 2006;5:375–9. doi: 10.4161/cbt.5.4.2522. [DOI] [PubMed] [Google Scholar]
  • 64.Akashi Y, Okamoto I, Iwasa T, Yoshida T, Suzuki M, Hatashita E, et al. Enhancement of the antitumor activity of ionising radiation by nimotuzumab, a humanised monoclonal antibody to the epidermal growth factor receptor, in non-small cell lung cancer cell lines of differing epidermal growth factor receptor status. Br J Cancer. 2008;98:749–55. doi: 10.1038/sj.bjc.6604222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Navarro BG, Parada AC, Alvarez P, Leon A, Santana E, Bada A, et al. Local and systemic toxicity of h-R3, an anti-epidermal growth factor receptor monoclonal antibody, labeled with 188osmiun after the intracerebral administration in rats. Exp Toxicol Pathol. 2005;56:313–9. doi: 10.1016/j.etp.2004.07.004. [DOI] [PubMed] [Google Scholar]
  • 66.Casacó A, Lopez G, Garcia I, Rodriguez JA, Fernandez R, Figueredo J, et al. Phase I single-dose study of intracavitary-administered Nimotuzumab labeled with 188 Re in adult recurrent high-grade glioma. Cancer Biol Ther. 2008;7:333–9. doi: 10.4161/cbt.7.3.5414. [DOI] [PubMed] [Google Scholar]
  • 67.Torres LA, Perera A, Batista JF, Hernandez A, Crombet T, Ramos M, et al. Phase I/II clinical trial of the humanized anti-EGF-r monoclonal antibody h-R3 labelled with 99mTc in patients with tumour of epithelial origin. Nucl Med Commun. 2005;26:1049–57. doi: 10.1097/00006231-200512000-00002. [DOI] [PubMed] [Google Scholar]
  • 68.Vera DR, Eigner S, Beran M, Henke KE, Laznickova A, Laznicek M, et al. Preclinical evaluation of (177)lu-nimotuzumab: a potential tool for radioimmunotherapy of epidermal growth factor receptor-overexpressing tumors. Cancer Biother Radiopharm. 2011;26:287–97. doi: 10.1089/cbr.2010.0916. [DOI] [PubMed] [Google Scholar]
  • 69.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. [PubMed] [Google Scholar]
  • 70.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. doi: 10.1038/nrd1381. [DOI] [PubMed] [Google Scholar]
  • 71.Hsu JY, Wakelee HA. Monoclonal antibodies targeting vascular endothelial growth factor: current status and future challenges in cancer therapy. BioDrugs. 2009;23:289–304. doi: 10.2165/11317600-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 72.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. doi: 10.1200/JCO.2009.23.7537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.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. doi: 10.1046/j.1365-2141.1998.00835.x. [DOI] [PubMed] [Google Scholar]
  • 74.Cavarretta IT, Neuwirt H, Untergasser G, Moser PL, Zaki MH, Steiner H, et al. The antiapoptotic effect of IL-6 autocrine loop in a cellular model of advanced prostate cancer is mediated by Mcl-1. Oncogene. 2007;26:2822–32. doi: 10.1038/sj.onc.1210097. [DOI] [PubMed] [Google Scholar]
  • 75.Cavarretta IT, Neuwirt H, Zaki MH, Steiner H, Hobisch A, Nemeth JA, et al. Mcl-1 is regulated by IL-6 and mediates the survival activity of the cytokine in a model of late stage prostate carcinoma. Adv Exp Med Biol. 2008;617:547–55. doi: 10.1007/978-0-387-69080-3_56. [DOI] [PubMed] [Google Scholar]
  • 76.Dorff TB, Goldman B, Pinski JK, Mack PC, Lara PN, Jr., Van Veldhuizen PJ, Jr., et al. Clinical and correlative results of SWOG S0354: a phase II trial of CNTO328 (siltuximab), a monoclonal antibody against interleukin-6, in chemotherapy-pretreated patients with castration-resistant prostate cancer. Clin Cancer Res. 2010;16:3028–34. doi: 10.1158/1078-0432.CCR-09-3122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rossi JF, Negrier S, James ND, Kocak I, Hawkins R, Davis H, et al. A phase I/II study of siltuximab (CNTO 328), an anti-interleukin-6 monoclonal antibody, in metastatic renal cell cancer. Br J Cancer. 2010;103:1154–62. doi: 10.1038/sj.bjc.6605872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Puchalski T, Prabhakar U, Jiao Q, Berns B, Davis HM. Pharmacokinetic and pharmacodynamic modeling of an anti-interleukin-6 chimeric monoclonal antibody (siltuximab) in patients with metastatic renal cell carcinoma. Clin Cancer Res. 2010;16:1652–61. doi: 10.1158/1078-0432.CCR-09-2581. [DOI] [PubMed] [Google Scholar]
  • 79.van Rhee F, Fayad L, Voorhees P, Furman R, Lonial S, Borghaei H, et al. Siltuximab, a novel anti-interleukin-6 monoclonal antibody, for Castleman's disease. J Clin Oncol. 2010;28:3701–8. doi: 10.1200/JCO.2009.27.2377. [DOI] [PubMed] [Google Scholar]
  • 80.Steiner H, Cavarretta IT, Moser PL, Berger AP, Bektic J, Dietrich H, et al. Regulation of growth of prostate cancer cells selected in the presence of interleukin-6 by the anti-interleukin-6 antibody CNTO 328. Prostate. 2006;66:1744–52. doi: 10.1002/pros.20492. [DOI] [PubMed] [Google Scholar]
  • 81.Voorhees PM, Chen Q, Kuhn DJ, Small GW, Hunsucker SA, Strader JS, et al. Inhibition of interleukin-6 signaling with CNTO 328 enhances the activity of bortezomib in preclinical models of multiple myeloma. Clin Cancer Res. 2007;13:6469–78. doi: 10.1158/1078-0432.CCR-07-1293. [DOI] [PubMed] [Google Scholar]
  • 82.Voorhees PM, Chen Q, Small GW, Kuhn DJ, Hunsucker SA, Nemeth JA, et al. Targeted inhibition of interleukin-6 with CNTO 328 sensitizes pre-clinical models of multiple myeloma to dexamethasone-mediated cell death. Br J Haematol. 2009;145:481–90. doi: 10.1111/j.1365-2141.2009.07647.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hunsucker SA, Magarotto V, Kuhn DJ, Kornblau SM, Wang M, Weber DM, et al. Blockade of interleukin-6 signalling with siltuximab enhances melphalan cytotoxicity in preclinical models of multiple myeloma. Br J Haematol. 2011;152:579–92. doi: 10.1111/j.1365-2141.2010.08533.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Song L, Rawal B, Nemeth JA, Haura EB. JAK1 activates STAT3 activity in non-small-cell lung cancer cells and IL-6 neutralizing antibodies can suppress JAK1-STAT3 signaling. Mol Cancer Ther. 2011;10:481–94. doi: 10.1158/1535-7163.MCT-10-0502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.McKian KP, Haluska P. Cixutumumab. Expert Opin Investig Drugs. 2009;18:1025–33. doi: 10.1517/13543780903055049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Scartozzi M, Bianconi M, Maccaroni E, Giampieri R, Berardi R, Cascinu S. Dalotuzumab, a recombinant humanized mAb targeted against IGFR1 for the treatment of cancer. Curr Opin Mol Ther. 2010;12:361–71. [PubMed] [Google Scholar]
  • 87.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. doi: 10.1158/1078-0432.CCR-04-1645. [DOI] [PubMed] [Google Scholar]
  • 88.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. 2011 doi: 10.1038/cdd.2011.96. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.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. doi: 10.1038/cdd.2008.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ran S, Downes A, Thorpe PE. Increased exposure of anionic phospholipids on the surface of tumor blood vessels. Cancer Res. 2002;62:6132–40. [PubMed] [Google Scholar]
  • 91.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. doi: 10.1016/S0360-3016(02)03928-7. [DOI] [PubMed] [Google Scholar]
  • 92.Dolloff NG, Russell MR, Loizos N, Fatatis A. Human bone marrow activates the Akt pathway in metastatic prostate cells through transactivation of the alpha-platelet-derived growth factor receptor. Cancer Res. 2007;67:555–62. doi: 10.1158/0008-5472.CAN-06-2593. [DOI] [PubMed] [Google Scholar]
  • 93.Shah GD, Loizos N, Youssoufian H, Schwartz JD, Rowinsky EK. Rationale for the development of IMC-3G3, a fully human immunoglobulin G subclass 1 monoclonal antibody targeting the platelet-derived growth factor receptor alpha. Cancer. 2010;116:1018–26. doi: 10.1002/cncr.24788. [DOI] [PubMed] [Google Scholar]
  • 94.Russell MR, Liu Q, Fatatis A. Targeting the {alpha} receptor for platelet-derived growth factor as a primary or combination therapy in a preclinical model of prostate cancer skeletal metastasis. Clin Cancer Res. 2010;16:5002–10. doi: 10.1158/1078-0432.CCR-10-1863. [DOI] [PubMed] [Google Scholar]
  • 95.Ribas A, Camacho LH, Lopez-Berestein G, Pavlov D, Bulanhagui CA, Millham R, et al. Antitumor activity in melanoma and anti-self responses in a phase I trial with the anti-cytotoxic T lymphocyte-associated antigen 4 monoclonal antibody CP-675,206. J Clin Oncol. 2005;23:8968–77. doi: 10.1200/JCO.2005.01.109. [DOI] [PubMed] [Google Scholar]
  • 96.Calabrò L, Danielli R, Sigalotti L, Maio M. Clinical studies with anti-CTLA-4 antibodies in non-melanoma indications. Semin Oncol. 2010;37:460–7. doi: 10.1053/j.seminoncol.2010.09.006. [DOI] [PubMed] [Google Scholar]
  • 97.Camacho LH, Antonia S, Sosman J, Kirkwood JM, Gajewski TF, Redman B, et al. Phase I/II trial of tremelimumab in patients with metastatic melanoma. J Clin Oncol. 2009;27:1075–81. doi: 10.1200/JCO.2008.19.2435. [DOI] [PubMed] [Google Scholar]
  • 98.Rini BI, Stein M, Shannon P, Eddy S, Tyler A, Stephenson JJ, Jr., et al. Phase 1 dose-escalation trial of tremelimumab plus sunitinib in patients with metastatic renal cell carcinoma. Cancer. 2011;117:758–67. doi: 10.1002/cncr.25639. [DOI] [PubMed] [Google Scholar]
  • 99.Oldenhuis CN, Stegehuis JH, Walenkamp AM, de Jong S, de Vries EG. Targeting TRAIL death receptors. Curr Opin Pharmacol. 2008;8:433–9. doi: 10.1016/j.coph.2008.06.011. [DOI] [PubMed] [Google Scholar]
  • 100.Doi T, Murakami H, Ohtsu A, Fuse N, Yoshino T, Yamamoto N, et al. Phase 1 study of conatumumab, a pro-apoptotic death receptor 5 agonist antibody, in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol. 2011;68:733–41. doi: 10.1007/s00280-010-1544-1. [DOI] [PubMed] [Google Scholar]
  • 101.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. doi: 10.1093/annonc/mdn015. [DOI] [PubMed] [Google Scholar]
  • 102.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. [PubMed] [Google Scholar]
  • 103.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. doi: 10.1089/cbr.2009.0673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Herbst RS, Kurzrock R, Hong DS, Valdivieso M, Hsu CP, Goyal L, et al. A first-in-human study of conatumumab in adult patients with advanced solid tumors. Clin Cancer Res. 2010;16:5883–91. doi: 10.1158/1078-0432.CCR-10-0631. [DOI] [PubMed] [Google Scholar]
  • 105.Rajeshkumar NV, Rasheed ZA, Garcia-Garcia E, Lopez-Rios F, Fujiwara K, Matsui WH, et al. A combination of DR5 agonistic monoclonal antibody with gemcitabine targets pancreatic cancer stem cells and results in long-term disease control in human pancreatic cancer model. Mol Cancer Ther. 2010;9:2582–92. doi: 10.1158/1535-7163.MCT-10-0370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.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. [PubMed] [Google Scholar]
  • 107.Wang W, Lau R, Yu D, Zhu W, Korman A, Weber J. PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi) regulatory T cells. Int Immunol. 2009;21:1065–77. doi: 10.1093/intimm/dxp072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–75. doi: 10.1200/JCO.2009.26.7609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Haas C, Krinner E, Brischwein K, Hoffmann P, Lutterbuse R, Schlereth B, et al. Mode of cytotoxic action of T cell-engaging BiTE antibody MT110. Immunobiology. 2009;214:441–53. doi: 10.1016/j.imbio.2008.11.014. [DOI] [PubMed] [Google Scholar]
  • 110.Nagorsen D, Baeuerle PA. Immunomodulatory therapy of cancer with T cell-engaging BiTE antibody blinatumomab. Exp Cell Res. 2011;317:1255–60. doi: 10.1016/j.yexcr.2011.03.010. [DOI] [PubMed] [Google Scholar]
  • 111.Sharkey RM, Rossi EA, McBride WJ, Chang CH, Goldenberg DM. Recombinant bispecific monoclonal antibodies prepared by the dock-and-lock strategy for pretargeted radioimmunotherapy. Semin Nucl Med. 2010;40:190–203. doi: 10.1053/j.semnuclmed.2009.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Schoffelen R, van der Graaf WT, Franssen G, Sharkey RM, Goldenberg DM, McBride WJ, et al. Pretargeted 177Lu radioimmunotherapy of carcinoembryonic antigen-expressing human colonic tumors in mice. J Nucl Med. 2010;51:1780–7. doi: 10.2967/jnumed.110.079376. [DOI] [PubMed] [Google Scholar]
  • 113.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. doi: 10.1182/blood-2009-02-205476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Vallera DA, Elson M, Brechbiel MW, Dusenbery KE, Burns LJ, Skubitz KM, et al. Preclinical studies targeting normal and leukemic hematopoietic cells with Yttrium-90-labeled anti-CD45 antibody in vitro and in vivo in nude mice. Cancer Biother Radiopharm. 2003;18:133–45. doi: 10.1089/108497803765036300. [DOI] [PubMed] [Google Scholar]
  • 115.Vallera DA, Sicheneder AR, Taras EP, Brechbiel MW, Vallera JA, Panoskaltsis-Mortari A, et al. Radiotherapy of CD45-expressing Daudi tumors in nude mice with yttrium-90-labeled, PEGylated anti-CD45 antibody. Cancer Biother Radiopharm. 2007;22:488–500. doi: 10.1089/cbr.2007.366. [DOI] [PubMed] [Google Scholar]
  • 116.Wong JY, Chu DZ, Williams LE, Liu A, Zhan J, Yamauchi DM, et al. A phase I trial of (90)Y-DOTA-anti-CEA chimeric T84.66 (cT84.66) radioimmunotherapy in patients with metastatic CEA-producing malignancies. Cancer Biother Radiopharm. 2006;21:88–100. doi: 10.1089/cbr.2006.21.88. [DOI] [PubMed] [Google Scholar]
  • 117.Yazaki PJ, Sherman MA, Shively JE, Ikle D, Williams LE, Wong JY, et al. Humanization of the anti-CEA T84.66 antibody based on crystal structure data. Protein Eng Des Sel. 2004;17:481–9. doi: 10.1093/protein/gzh056. [DOI] [PubMed] [Google Scholar]
  • 118.Jurcic J. Ab therapy of AML: native anti-CD33 Ab and drug conjugates. Cytotherapy. 2007;4:1–6. doi: 10.1080/14653240701519012. [DOI] [PubMed] [Google Scholar]
  • 119.Feldman EJ, Brandwein J, Stone R, Kalaycio M, Moore J, O'Connor J, et al. Phase III randomized multicenter study of a humanized anti-CD33 monoclonal antibody, lintuzumab, in combination with chemotherapy, versus chemotherapy alone in patients with refractory or first-relapsed acute myeloid leukemia. J Clin Oncol. 2005;23:4110–6. doi: 10.1200/JCO.2005.09.133. [DOI] [PubMed] [Google Scholar]
  • 120.Gibson AD. Phase III trial of a humanized anti-CD33 antibody (HuM195) in patients with relapsed or refractory acute myeloid leukemia. Clin Lymphoma. 2002;3:18–9. doi: 10.1016/s1526-9655(11)70245-7. [DOI] [PubMed] [Google Scholar]
  • 121.Abès R, Gelize E, Fridman WH, Teillaud JL. Long-lasting antitumor protection by anti-CD20 antibody through cellular immune response. Blood. 2010;116:926–34. doi: 10.1182/blood-2009-10-248609. [DOI] [PubMed] [Google Scholar]
  • 122.Kim PS, Armstrong TD, Song H, Wolpoe ME, Weiss V, Manning EA, et al. Antibody association with HER-2/neu-targeted vaccine enhances CD8 T cell responses in mice through Fc-mediated activation of DCs. J Clin Invest. 2008;118:1700–11. doi: 10.1172/JCI34333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Taylor C, Hershman D, Shah N, Suciu-Foca N, Petrylak DP, Taub R, et al. Augmented HER-2 specific immunity during treatment with trastuzumab and chemotherapy. Clin Cancer Res. 2007;13:5133–43. doi: 10.1158/1078-0432.CCR-07-0507. [DOI] [PubMed] [Google Scholar]
  • 124.Park S, Jiang Z, Mortenson ED, Deng L, Radkevich-Brown O, Yang X, et al. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell. 2010;18:160–70. doi: 10.1016/j.ccr.2010.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Stagg J, Sharkey J, Pommey S, Young R, Takeda K, Yagita H, et al. Antibodies targeted to TRAIL receptor-2 and ErbB-2 synergize in vivo and induce an antitumor immune response. Proc Natl Acad Sci USA. 2008;105:16254–9. doi: 10.1073/pnas.0806849105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Abès R, Teillaud JL. Modulation of tumor immunity by therapeutic monoclonal antibodies. Cancer Metastasis Rev. 2011;30:111–24. doi: 10.1007/s10555-011-9282-3. [DOI] [PubMed] [Google Scholar]
  • 127.Harbers SO, Crocker A, Catalano G, D'Agati V, Jung S, Desai DD, et al. Antibody-enhanced cross-presentation of self antigen breaks T cell tolerance. J Clin Invest. 2007;117:1361–9. doi: 10.1172/JCI29470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Rafiq K, Bergtold A, Clynes R. Immune complex-mediated antigen presentation induces tumor immunity. J Clin Invest. 2002;110:71–9. doi: 10.1172/JCI15640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.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. doi: 10.1007/s10555-011-9273-4. [DOI] [PubMed] [Google Scholar]
  • 130.Wilson NS, Yang B, Yang A, Loeser S, Marsters S, Lawrence D, et al. An Fcgamma receptor-dependent mechanism drives antibody-mediated target-receptor signaling in cancer cells. Cancer Cell. 2011;19:101–13. doi: 10.1016/j.ccr.2010.11.012. [DOI] [PubMed] [Google Scholar]
  • 131.Cassard L, Cohen-Solal JF, Fournier EM, Camilleri-Broet S, Spatz A, Chouaib S, et al. Selective expression of inhibitory Fcgamma receptor by metastatic melanoma impairs tumor susceptibility to IgG-dependent cellular response. Int J Cancer. 2008;123:2832–9. doi: 10.1002/ijc.23870. [DOI] [PubMed] [Google Scholar]
  • 132.Cassard L, Cohen-Solal JF, Galinha A, Sastre-Garau X, Mathiot C, Galon J, et al. Modulation of tumor growth by inhibitory Fc(gamma) receptor expressed by human melanoma cells. J Clin Invest. 2002;110:1549–57. doi: 10.1172/JCI15454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Rascu A, Repp R, Westerdaal NA, Kalden JR, van de Winkel JG. Clinical relevance of Fc gamma receptor polymorphisms. Ann N Y Acad Sci. 1997;815:282–95. doi: 10.1111/j.1749-6632.1997.tb52070.x. [DOI] [PubMed] [Google Scholar]
  • 134.Zhang W, Gordon M, Schultheis AM, Yang DY, Nagashima F, Azuma M, et al. FCGR2A and FCGR3A polymorphisms associated with clinical outcome of epidermal growth factor receptor expressing metastatic colorectal cancer patients treated with single-agent cetuximab. J Clin Oncol. 2007;25:3712–8. doi: 10.1200/JCO.2006.08.8021. [DOI] [PubMed] [Google Scholar]
  • 135.Paiva M, Marques H, Martins A, Ferreira P, Catarino R, Medeiros R. FcgammaRIIa polymorphism and clinical response to rituximab in non-Hodgkin lymphoma patients. Cancer Genet Cytogenet. 2008;183:35–40. doi: 10.1016/j.cancergencyto.2008.02.001. [DOI] [PubMed] [Google Scholar]
  • 136.Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood. 2002;99:754–8. doi: 10.1182/blood.V99.3.754. [DOI] [PubMed] [Google Scholar]
  • 137.de Romeuf C, Dutertre CA, Le Garff-Tavernier M, Fournier N, Gaucher C, Glacet A, et al. Chronic lymphocytic leukaemia cells are efficiently killed by an anti-CD20 monoclonal antibody selected for improved engagement of FcgammaRIIIA/CD16. Br J Haematol. 2008;140:635–43. doi: 10.1111/j.1365-2141.2007.06974.x. [DOI] [PubMed] [Google Scholar]

Articles from Oncoimmunology are provided here courtesy of Taylor & Francis

RESOURCES