Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Aug 14.
Published in final edited form as: Expert Opin Biol Ther. 2013 Jun 26;13(8):1187–1196. doi: 10.1517/14712598.2013.810717

Targeted inhibition of VEGF Receptor-2: An update on Ramucirumab

JM Clarke 1, HI Hurwitz 1
PMCID: PMC4131847  NIHMSID: NIHMS599912  PMID: 23803182

Abstract

Introduction

Ramucirumab (IMC-1121B) is a fully humanized IgG1 monoclonal antibody targeting the extracellular domain of VEGF receptor 2 (VEGFR2). Numerous phase I-II trials with ramucirumab in various malignancies have shown promising clinical antitumor efficacy and tolerability. Most recently, the large phase III REGARD trial evaluated ramucirumab in patients with refractory metastatic gastric cancer. Patients receiving ramucirumab experienced a median overall survival of 5.2 months compared to 3.8 months on placebo.

Areas Covered

The purpose of this article is to review the preclinical motivation for VEGFR2 targeted therapies and survey recent data from clinical trials involving ramucirumab, as well as highlight ongoing studies.

Expert Opionion

Rational multi-target approaches to angiogenesis are needed to overcome resistance mechanisms. Predictive angiogenic biomarkers are also needed to optimize patient selection for novel anti-angiogenic agents.

Keywords: Ramucirumab, angiogenesis, cancer, VEGF, VEGFR2, monoclonal antibody, gastric cancer, adenocarcinoma, vascular endothelial growth factor

1.1 Introduction

Sustained angiogenesis is a hallmark of cancer; and targeted inhibition of blood vessel development is an established modality of antitumor therapy1. During the past decade, multiple clinical trials have demonstrated improved survival by inhibiting angiogenesis, principally the vascular endothelial growth factor (VEGF) axis, by one of two mechanisms: monoclonal antibody binding of VEGFA [VEGF] with bevacizumab and multi-target receptor tyrosine kinase (RTK) inhibitors with anti-angiogenic specificity. Anti-VEGF therapies have been associated with a survival benefit across multiple malignancies including colorectal, non-small cell lung (NSCLC), renal cell, and hepatocellular carcinomas, among others2-5. Accordingly, novel approaches resulting in greater blockade of VEGF signaling and inhibition of angiogenesis have generated substantial interest.

Ramucirumab (IMC-1121B) is a fully humanized IgG1 monoclonal antibody targeting the extracellular domain of VEGF receptor 2 (VEGFR2). VEGFR2 is largely considered the primary VEGF family receptor driving angiogenesis. While multiple lines of evidence support the preclinical efficacy of targeted VEGFR2 inhibition, within the past several years, numerous phase I-II trials have showed promising clinical antitumor effect and tolerability. Most recently, the results of the large phase III REGARD trial of ramucirumab in patients with refractory metastatic gastric cancer were announced. The primary aim of this article is to review the preclinical motivation for VEGFR2 targeted therapies and survey recent data from clinical trials involving ramucirumab.

1.2 VEGF signaling and Tumor Angiogenesis

Tumor angiogenesis is a highly complex process dependent on a multi-faceted program of endothelial cell activation, stromal cell and endothelial-progenitor recruitment, extra-cellular matrix remodeling, pro-angiogenic cytokine signaling, and activation of oncogenic signaling cascades6, 7. A critical pillar of angiogenesis is the interaction of the VEGF family of pro-angiogenic cytokines and their respective receptors. The VEGF family includes VEGFA, VEGFB, VEGFC, VEGFD, VEGFE, and placental growth factor (PlGF) with three receptors: VEGFR1 (fms-like tyrosine kinase 1/Flt-1), VEGFR2 (Flk-1/KDR), and VEGFR3 (Flt-4) with associated co-reptors neuropillin 1 and 2 (NRP1/2)(see fig. 1)8. VEGFR2 expression is typically limited to vessel endothelial cells, while VEGFR1 can also be found on bone-marrow derived progenitors9. VEGFR1 classically binds to homodimers of VEGFA, VEGFB, and PlGF, while VEGFR2 binds VEGFA, VEGFE and processed forms of VEGFC and VEGFD10. While VEGFR3 is principally involved in lympangiogenesis, VEGFR2 is widely considered the main receptor driving angiogenesis. VEGFR1 exhibits high binding affinity for VEGFA, but weak phosphorylation activity possibly suggesting a negative modulatory role on VEGF signaling9, 11. Furthermore, both PlGF and VEGFB bind exclusively to VEGFR1, can form heterodimers with VEGFA, and affect tumor growth12-16. VEGFR1 is also implicated in monocyte chemotaxis, hematopoietic stem cell (HSC) survival, and inhibition of dendritic cell maturation17-20, while soluble VEGFR1 plays a role in pre-eclampsia21. Upregulation of VEGFA mRNA is demonstrated across almost every type malignancy, including breast, lung, colorectal cancers, renal cell, ovarian, and glioblastoma22. Likewise, upregulation of VEGFR2 expression is seen in the tumor vasculature in a variety of malignancies as well 23-25.

Figure 1.

Figure 1

Binding of VEGFR2 to its ligand results in intracellular transphosphorylation and activation of multiple downstream pathways including PLC-γ, MAPK, PI3K, Akt, and Src. Proangiogenic signals and VEGFR activity is modulated by several receptors including integrins, FGFR, PDGFR, Notch, and TIE2. Activation of VEGFR1 leads to downstream effects on HSC, DC maturation, and chemotaxis, while VEGFR3 promotes lymphangiogenesis. General drug targets are illustrated for aflibercept, bevacizumab, ramucirumab, and multiple RTK inhibitors.

The VEGFRs belong to the immunoglobulin subclass of the receptor tyrosine kinase (RTK) superfamily and have seven Ig-like extracellular domains with a single transmembrane helix with an intracellular kinase region10, 26. Following binding of VEGFA, VEGFR2 has the potential to form either a homodimer or heterodimer complex with VEGFR1, resulting in intracellular tyrosine phosphorylation10, 26. Ultimately, downstream effects of VEGFR2 signaling which culminate in angiogenesis, include a potent increase in vascular permeability and promotion of endothelial cell migration, proliferation, and survival10. Complicating the differential effects of VEGFA signaling and VEGFR2 activation, is the substantial crosstalk with other pro-angiogenic molecules and adhesion proteins, including multiple VEGF isoforms, angiopoetins, and integrins6. Ang/Tie, Dll4/Notch, and av integrin signaling pathways all intersect with the VEGF axis and modulate angiogenesis, lymph-angiogenesis, and metastasis (see fig. 1)6, 27.

Binding of VEGFs to VEGFR2 initiates receptor dimerization and robust intracellular autophosphorylation of multiple tyrosine residues with numerous downstream consequences. Specifically, phosphorylation of Y1175 allows docking of phospholipase C-gamma (PLC-γ) resulting in activation of the mitogen activated protein kinase (MAPK) pathway and promotion of endothelial cell proliferation9, 10. Furthermore, Y1175 mediates phosphatidylinositol 3′ kinase (PI3K) activity leading, ultimately, to increased cell survival through AKT/PKB, cell migration, and vascular permeability via expression of endothelial nitric oxide synthase. Phosphorylation of other critical residues include Y951 and Y1214, which also promote vascular permeability, actin remodeling, and cell migration by way of Src/TSAd and P38/MAPK pathways9, 10.

Disruption of VEGFR2 signaling by currently FDA approved, anti-angiogenic agents occurs namely by either specific binding of circulating VEGF or small molecule inhibition of RTKs. Bevacizumab potently binds VEGFA reventing its docking with VEGFR1 and VEGFR2. Bevacizumab is widely used in mCRC, nonsmall cell lung, glioblastoma, and renal cell carcinomas. More recently, ziv-Afliberacept, a soluble VEGF receptor decoy with VEGFA, VEGFB, and placenta growth factor (PlGF) affinity, demonstrated efficacy in treatment refractory metastatic colorectal cancer (mCRC) patients, but not in NSCLC, prostate, or pancreatic adenocarcinoma 28-31. The number of FDA approved small molecule RTK inhibitors with anti-angiogenic specificity has increased significantly within the past several years. Currently, there are seven FDA approved mutlikinase inhibitors that are known to target VEGF (alone or with other targets):sorafenib, sunitinib, axitinib, pazopanib, vandetinib, cabozantinib, and regorafenib. Many of these agents demonstrate VEGFR inhibition, along with blockade of other receptors, such as PDGF and cKit. However, off target promiscuity coupled with incomplete blockade of pro-angiogenic kinases can theoretically cause RTK inhibitors to be associated with toxicity and have suboptimal antitumor activity32. Thus, ramucirumab offers a novel mechanism for anti-angiogenic therapy with the potential for both high affinity and high specificity blockade of VEGFR2,

2. Introduction to Ramucirumab

2.1 Preclinical Evidence

Due to species-specific differences in human VEGFR2 (KDR) and murine VEGFR2 (flk-1), the development of anti-VEGFR2 antibodies has required the production of immunoglobulins specific to both the human and murine forms of the receptor to sterically block ligand binding. In 1998, Witte et al initially described the development anti-flk-1 (DC101) and anti-KDR (p1C11) high affinity monoclonal antibodies, which demonstrated in vitropotent inhibition of VEGF receptor binding, intracellular phosphorylation and signaling, and human umbilical vein endothelial cell (HUVEC) mitogenesis33. DC101 was later shown, by the same group, to suppress the growth of primary murine lung, mammary, melanoma in vivo and inhibited multiple other human tumor xenografts34. DC101 effects included tumor cell apoptosis, decreased vessel density, and reduced tumor cell proliferation.

In 2003, Lu et al used a large phage display library with tailored in vitroselection methods to identify a high affinity antibody, 1121, with a >30-fold higher binding affinity to KDR compared to other candidate VEGFR2 antibodies. 1121 blocked VEGFA/KDR interaction with an IC50 of 1 nM and potently inhibited VEGF-stimulated KDR phosphorylation35. More recently, Miao et al in 2006 reported the production of a humanized anti-KDR Fab fragment leading to the generation of Fab 1121B, which retained high affinity for KDR. Indeed, 1121B was subsequently shown to block VEGFA binding, neutralize VEGFA-stimulated phosphorylation of KDR, and inhibit HUVEC mitogenesis36.

2.2 Chemistry

Ramucirumab is a fully humanized immunoglobulin G1 monoclonal antibody37. During initial preclinical development, a conserved variable heavy chain (VH) sequence was identified between multiple potential parent compounds, with reported greatest homology to the germline DP77 segment of the human VH3 family. This single VH was recombined with variable light chains (VL) using a phage display library with subsequent rounds of affinity maturation selection. The resulting consensus VH/VL combination was labeled as 1121, the amino acid sequence of which has been previously reported35.

2.3 Pharmacodynamics and Pharmacokinetics

Based on preclinical in vitro data, the binding affinity of the 1121B Fab to KDR demonstrated an ED50 of approximately 0.1-0.15 nM. VEGFA, the primary native ligand for VEGFR2 has an affinity to VEGFR2 of .77-.88 nM, or approximately 8-9 fold weaker than the 1121B monoclonal antibody35, 36. 1121B effectively binds KDR both as a soluble protein and as a cell-surface based receptor, with an IC50 of 1-2 nM36. A detailed crystal structure analysis of the 1121B:KDR complex was performed by Franklin et al in 2011 showing that 1121B Fab binds to domain 3 of KDR near the N-terminus38. The epitope for 1121B binding consists of a B-hairpin with an adjacent B-strand, and domain 3 of the KDR receptor. Inhibition of VEGFA binding to KDR is likely mediated by both steric blocking of the ligand and induction of conformation change in the receptor when in contact with 1121B38.

In the initial phase I study of ramucirumab, a total of 37 patients were treated with doses ranging from 2 to 16 mg/kg infused weekly37. Favorable pharmacokinetic data was obtained from the study, as all patients demonstrated trough levels greater than the target of 20 ug/mL, and the half-life at steady-state ranged at 200-300 hours for 8-16 mg/kg doses. A nonlinear effect of the ramucirumab dose was seen on the clearance rate suggesting saturation of the clearance mechanism, which was likely to be largely receptor-mediated. However, minimal serum drug accumulation was evident over the course of the study. Despite large inter-patient variability, the findings were consistent with PK data from other anti-receptor antibodies37.

Pharmacodynamic data from the phase I clinical trial incorporated serum measurement of VEGFA and soluble VEGFR1/2 at time points before and during each cycle of treatment37. Following the first infusion, an immediate increased in VEGF of 1.5-3 fold over the pretreatment level was measured, which lasted the duration of the treatment course. VEGFR1/2 levels immediately decreased after the initial infusion of ramucirumab, then returned to baseline levels. Neither the VEGF or VEGFR1/2 change was dose related, suggesting saturation of the receptor as also described by the PK data. Sequential DCE-MRI measurement did confirm reduced tumor vascularity in 69% of the patients. Importantly, no anti-ramucirumab antibodies were detected at the conclusion of treatment in any of the patients37.

3. Clinical Evidence using Ramucirumab

3.1 Phase I and II Trials

Two phase I studies with ramucirumab have been completed to date, however the results of only one trial have been fully published37, 39. Spratlin et al in 2010 reported the phase I results with ramucirumab in 37 patients with advanced solid malignancies. The majority of the patients had received prior chemotherapy, however less than 15% had reported prior exposure to anti-angiogenic therapies. A standard 3+3 dose escalation scheme was used with weekly administration of ramucirumab starting at 2 mg/kg. Patients were treated up to 16 mg/kg, however 2 patients developed dose-limiting hypertension and venous thrombosis, thus 13 mg/kg was determined to be the maximum tolerated dose. 60% of patients developed grade 3 or higher toxicity with fatigue, nausea/vomiting, proteinuria, and hypertension being noted. Promising efficacy was observed as 4 of 27 patients with measurable disease had a partial response. Partial response or stable disease was seen in 73% of patients, and 11 of 37 patients had a partial response or stable disease at 6 months follow up. A smaller study in 2007 also evaluated q2 week or q3 week dosing regimens with ramucirumab and demonstrated similar PK and safety data39.

Given the promising phase I results, numerous phase II disease specific trials have been performed over the past five years. Beginning in 2010, results of combinations of ramucirumab with various chemotherapy regimens were reported in metastatic melanoma, metastatic renal cell carcinoma (RCC), NSCLC, and hepatocellular carcinoma (HCC). Many of these studies have been reviewed previously40. Insorafenib-naïve patients with advanced HCC, ramucirumab demonstrated a progression free survival of 4.3 months41. In advanced NSCLC patients, combination ramucirumab q3 weeks with carboplatin and paclitaxel demostrated an overall response rate of 67% with a progression free survival of 5. 7 months42. Additionally, in metastatic RCC patients with previous sorafenib/sunitinib exposure, ramucirumab showed a median progression free survival of 6 months with nearly 50% of patients having stable disease at >5 months. Finally, a randomized trial of patients with metastatic melanoma compared ramucirumab with or without dacarbazine. Although, treatment was relatively well tolerated, efficacy was poor with a progression free survival of 1.6 and 2.5 months, respectively, in each group43.

Within the past year, additional phase II study results have been presented in ovarian, prostate, and colorectal cancers. Ramucirumab was evaluated in women with persistent or recurrent epithelial ovarian, fallopian tube, or primary peritoneal carcinoma after previous platinum- based chemotherapy. Approximately one third of patients had progression free survival at 6 months, with a median progression free survival of 3.5 months and OS of 11.1 months44. Efficacy of ramucirumab in metastatic castrate resistant prostate cancer (CRPC) was studied in a randomized phase II study comparing mitoxantrone / prednisone with either ramucirumab or cixutumumab45. Patients were randomized between treatment arms. PSA response was seen in 22%, with a progression free survival and overall survival of 6.7 months and 13.0 months, respectively. In metastatic colorectal cancer, ramucirumab was combined with FOLFOX chemotherapy; this regimen demonstrated a median progression free survival of 11.5 months, with an ORR of 67%, including 5 patients with complete response and 27 with a partial response46.

Multiple studies are currently ongoing evaluating novel combinations of ramucirumab with chemotherapy. Vahdat et al are examining ramucirumab in a 3-arm trial in metastatic breast cancer patients previously treated with an anthracycline and taxane. Patients will be randomized to capecitabine alone or in combination with either IMC-18F1 (VEGFR1 inhibitor) or ramucirumab47. A similar 3 arm study is ongoing to evaluate ramucirumab or IMC-18F1 with docetaxel in advanced urothelial cell carcinoma following platinum-based chemotherapy48. Other studies evaluating ramucirumab with docetaxel in metastatic NSCLC and in combination with FOLFIRI in advanced solid tumors are also currently enrolling (NCT01703091, NCT01634555).

3.2 Phase III Trials

The results of the pivotal phase III randomized trial for ramucirumab in metastatic gastric cancer have now been presented in full detail, however publication is pending49, 50. The REGARD trial was an international, placebo-controlled, double-blind trial evaluating efficacy and safety of ramucirumab monotherapy in patients with metastatic gastric or gastroesophageal junction cancer following standard first line treatment with platinum or fluoropyrimidine based therapy. A total of 355 patients were randomized in a 2:1 fashion to 8 mg/kg or placebo every 2 weeks in combination with best supportive care. The study enrolled 355 patients from 30 countries at 120 different centers. The primary endpoint of the trial was overall survival, with secondary endpoints of progression free survival and quality of life. Following randomization, median age was 60 years with approximately 75% of patients with metastatic gastric cancer and 25% metastatic gastroesophageal junction cancer in each treatment arm. Patients were well matched with respect to number of sites of metastases, histologic subtype, presence of peritoneal disease, and response to prior therapy.

Patients receiving ramucirumab on average received 4 infusions over 8 weeks compared to 3 infusion over 6 weeks with placebo. Ramucirumab demonstrated improved overall survival (HR 0.78; p=0.047), with a median of 5.2 months compared to 3.8 months on placebo. Six- month and 12-month survival rates were 42% vs 32% and 18% vs 11%, respectively between ramucirumab and placebo arms. progression free survival was also significantly prolonged (HR=0.48), with median progression free survival of 1.3 months to 2.1 months with ramucirumab, as well as 12-week progression free survival (40% vs. 16%). While ORR was low between both arms (3.4% vs 2.6%), disease control rate (49% vs. 23%) was superior in the active treatment arm. Post-discontinuation treatment was performed in 31.5% of patients on ramucirumab compared with 39.3% on placebo. Sub-group analysis showed general consistent effects on overall survival and progression free survival independent of primary tumor location, preceding weight loss > 10%, geographic region, type of 1st line chemotherapy, and presence of peritoneal disease. Importantly, overall toxicity for patients in the ramucirumab arm was low (see Safety and Tolerability).

Given the favorable toxicity profile and survival benefit, ramucirumab is a potential new second line agent in metastatic gastric cancer. Prior to ramucirumab, antiangiogenic agents including bevacizumab, sunitinib and sorafenib have shown limited efficacy in 1st and 2nd line settings in metastatic gastric cancer51, 52. For instance, the recent AVAGAST trial of combination bevacizumab with cisplatin and capecitabine in 774 patients showed an improvement in progression free survival and response rate, but failed to meet its primary endpoint of overall survival53. Two recent phase III clinical trials have demonstrated a survival benefit of chemotherapy alone in the 2nd and 3rd line setting for patients with metastatic gastric cancer. Irinotecan demonstrated improved overall survival compared with best supportive care (BSC), HR of 0.48 (p=0. 012) with median survival of 4.0 months compared to 2.4 months54. Treatment with either irinotecan or docetaxel following progression after one or two prior chemotherapy regimens, yielded increased survival versus best supportive care with a HR of 0.657 (p=.007) and median overall survival of 5.2 months vs 3.8 months55. Based upon the activity of taxanes in 2nd line gastric cancer, the RAINBOW phase III trial is evaluating paclitaxel with or without ramucirumab for patients with metastatic treatment refractory gastric cancer (NCT01170663). Approximately 600 patients will be recruited from 200 study centers in 30 countries and results of this trial are expected in the near future.

Several other phase III trials of ramucirumab are currently in progress. A large study of ramucirumab versus placebo in patients with HCC following treatment with sorafenib is also ongoing. The primary endpoint is overall survival with goal of 544 patients powered to detect an increase of 2 months56. Additionally, a multinational randomized trial of docetaxel with or without ramucirumab for treatment naïve, HER2 negative, metastatic breast cancer patients is ongoing with a primary endpoint of progression free survival. Finally, FOLFIRI with or without ramucirumab will be evaluated in patients with metastatic CRC who have experience progression during 1st line treatment, including bevacizumab (NCT01183780).

3.3 Safety and Tolerability

The largest experience with safety and tolerability of targeted VEGFR2 inhibition is described by the REGARD trial50. In general ramucirumab was very well tolerated. Approximately 10% of patients discontinued ramucirumab treatment due to adverse event compare with 6.0% receiving placebo. Grade 3 or higher adverse events with rates that were higher in the the ramucirumab vs placebo arm included hypertension, abdominal pain, fatigue, and hyponatremia. VEGF class risks of special interest to ramucirumab were modestly increased compared to the control group and were generally in line with those seen with other VEGF inhibitors 57-59(see table 1). Combination chemotherapy with ramucirumab from phase II trials showed generally no significant unexpected toxicity. While dosing regimens varied between studies with either weekly or every 2-3 week infusions, toxicity rates seemed independent of dose or frequency of administration. Further phase III data with combination docetaxel or FOLFIRI, for example, will be helpful in delineating the toxicity profile with multi-drug regimens.

Table 1.

Comparison of therapies targeting VEGFR, with mechanism, and FDA approved indications.

Drug Bevacizumab ziv-Aflibercept Regorafenib Ramucirumab
Target(s) VEGFA VEGFA, VEGFB, PlGF VEGFR1-3, BRAF, PDGFR, KIT, RET, TIE2 VEGFR2
Mechanism Monoclonal antibody Fusion protein of VEGFR1 and VEGFR2 Multikinase inhibitor Monoclonal antibody
Dose 5-15 mg/kg IV Q2-3 weeks 4 mg/kg IV Q2 weeks 160 mg PO day 1-21 of 28 cycle 4-6 mg/kg Q2-3 weeks
FDA Approved indications Metastatic CRC, NSCLC, metastatic RCC, Glioblastoma Metastatic CRC Metastatic CRC NA

4. Conclusion

Ramucirumab is the first monoclonal antibody targeting VEGFR2 to be used in phase III clinical trials. The REGARD study demonstrated that ramucirumab is generally well tolerated and improves overall survival and progression free survival in refractory gastric and gastro-esophogeal cancers. Numerous additional phase II and III trials are currently ongoing to examine combination therapy of ramucirumab with cytotoxic chemotherapy in multiple disease settings, including 2nd line and 1st line metastatic gastric and gastro-esophogeal cancer, metastatic breast, NSCLC, colorectal, and HCC.

5. Expert Opinion

The demonstration of efficacy with targeted inhibition of VEGFR2 in a phase III trial represents an important milestone in anti-angiogenic therapy. However, the modest survival benefit illustrates a recurrent difficulty with anti-VEGF agents in clinical use. One explanation for the frequently varied clinical response and relatively transient benefit, may lie in the understanding of angiogenic resistance mechanisms. While VEGFR2 is considered the major signaling pathway of physiologic and pathological angiogenesis, a number of other pro-angiogenic pathways are known to contribute to tumor blood vessel formation including PDGF, FGF, angiopoietin, Ephrin, Dll4/Notch, and PlGF, among others60, 61. Inhibition of VEGFR2 (or VEGFA) may have some impact on these elements given pathway crosstalk, but is likely insufficient to prevent all escape mechanisms from occurring.

Despite these potential mechanisms of resistance, ramucirumab may have distinct mechanistic advantages compared to other anti-angiogenic modalities. For example, proteolytic processing of VEGFC and VEGFD allow their binding to VEGFR2 and promotion of angiogenesis, theoretically bypassing the effects of bevacizumab62. Of interest, in patients with mCRC treated with FOLFIRI and bevacizumab, VEGFC levels increased at time of disease progression63. Plasma VEGFD levels in pancreatic cancer patients were found to predict for benefit from bevacizumab when this agent was added to gemcitabine in the phase III study CALGB8030364. In the phase III MAX trial, tissue levels of VEGFD predicted for benefit from bevacizumab in combination with capecitabine and mitomycin C65. VEGFC and VEGFD signaling through VEGFR2, which may play a possible role in resistance to VEGFA targeted therapies, would be blocked with ramucirumab. The long half-life of ramucirumab would be expected to provide optimal VEGFR2 coverage, even if these ligands are up-regulated. However, VEGFA effects mediated via VEGFR1, would be blocked by bevacizumab or ziv-aflibercept, but not by ramicumerab. VEGFA can stimulate monocyte chemotaxis and inhibit dendritic cell maturation20, 66, 67. VEGFA can also induce proliferation, migration, and invasion of some tumor cells, at least in vitro16, 68. These effects on myeloid cells and tumor cells appear to be mediated by VEGFR1. Treatment with bevacizumab has been shown to reverse defective DC maturation and increase mature DCs in circulation67. Therefore, up-regulation of VEGFA as a result of VEGFR2 blockade by ramucirumab, may have effects on the immune system and angiogenesis by altering monocyte and dendritic cell function, and potentially even direct effects on the tumor cell.

Rational multi-target approaches to angiogenesis are needed to overcome resistance. The specificity, tolerability, and long half-life of ramucirumab suggest this agent will be well suited for combination anti-angiogenesis strategies. Predictive angiogenic biomarkers are also urgently needed to optimize patient selection, toxicity, and efficacy for the growing number of anti-angiogenic agents. Considerable data has been published and reviewed regarding the performance of various candidate predictive markers. Biomaker analyses, from REGARD and from other ramucirumab studies, are eagerly awaited.

Table 2.

Rates of selected adverse events observed in the REGARD trial, reported as percentages. Table adapted from Fuchs et al50.

Ramucirumab Placebo

Event Any Grade Grade ≥3 Any Grade Grade ≥3
Hypertension 16.1 7.6 7.8 2.6
Bleeding 12.7 3.4 11.3 2.6
Arterial thromboembolism 1.7 1.3 0 0
Venous thromboembolism 3.8 1.3 7 4.3
Proteinuria 3 0.4 2.6 0
GI perforation 0.8 0.8 0.9 0.9
Fistula 0.4 0.4 0.9 0.9
Infusion reaction 0.4 0 1.7 0
Heart Failure 0.4 0 0 0

Drug Summary Box.

Drug name (generic)

Ramucirumab

Phase (for indication under discussion)

Phase III

Indication (specific to discussion)

Metastatic gastric or gastroesophageal junction adenocarcinoma

Pharmacology description/mechanism of action

Vascular endothelial growth factor receptor-2 IgG1 monoclonal antibody

Route of administration

8 mg/kg intravenously every 2 weeks

Pivotal trial(s)

REGARD: A phase III, randomized, double-blinded trial of ramucirumab and best supportive care versus placebo and BSC in the treatment of metastatic gastric or gastroesophageal junction adenocarcinoma following disease progression on first-line platinum- and/or fluoropyrimidine-containing combination therapy.

References

  • 1.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000 Jan 7;100(1):57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
  • 2.Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004 Jun 3;350(23):2335–42. doi: 10.1056/NEJMoa032691. [DOI] [PubMed] [Google Scholar]
  • 3.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 Dec 14;355(24):2542–50. doi: 10.1056/NEJMoa061884. [DOI] [PubMed] [Google Scholar]
  • 4.Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007 Jan 11;356(2):115–24. doi: 10.1056/NEJMoa065044. [DOI] [PubMed] [Google Scholar]
  • 5.Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008 Jul 24;359(4):378–90. doi: 10.1056/NEJMoa0708857. [DOI] [PubMed] [Google Scholar]
  • 6.Weis SM, Cheresh DA. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med. 2011;17(11):1359–70. doi: 10.1038/nm.2537. [DOI] [PubMed] [Google Scholar]
  • 7.Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of angiogenesis. Cell. 2011 Sep 16;146(6):873–87. doi: 10.1016/j.cell.2011.08.039. [DOI] [PubMed] [Google Scholar]
  • 8.Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003 Jun;9(6):669–76. doi: 10.1038/nm0603-669. [DOI] [PubMed] [Google Scholar]
  • 9.Kowanetz M, Ferrara N. Vascular endothelial growth factor signaling pathways: therapeutic perspective. Clin Cancer Res. 2006 Sep 1;12(17):5018–22. doi: 10.1158/1078-0432.CCR-06-1520. [DOI] [PubMed] [Google Scholar]
  • 10.Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling - in control of vascular function. Nat Rev Mol Cell Biol. 2006 May;7(5):359–71. doi: 10.1038/nrm1911. [DOI] [PubMed] [Google Scholar]
  • 11.Kerbel RS. Tumor angiogenesis. N Engl J Med. 2008 May 8;358(19):2039–49. doi: 10.1056/NEJMra0706596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Yao J, Wu X, Zhuang G, Kasman IM, Vogt T, Phan V, et al. Expression of a functional VEGFR-1 in tumor cells is a major determinant of anti-PlGF antibodies efficacy. Proc Natl Acad Sci U S A. 2011 Jul 12;108(28):11590–5. doi: 10.1073/pnas.1109029108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hedlund EM, Yang X, Zhang Y, Yang Y, Shibuya M, Zhong W, et al. Tumor cell-derived placental growth factor sensitizes antiangiogenic and antitumor effects of anti-VEGF drugs. Proc Natl Acad Sci U S A. 2013 Jan 8;110(2):654–9. doi: 10.1073/pnas.1209310110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Albrecht I, Kopfstein L, Strittmatter K, Schomber T, Falkevall A, Hagberg CE, et al. Suppressive effects of vascular endothelial growth factor-B on tumor growth in a mouse model of pancreatic neuroendocrine tumorigenesis. PLoS One. 2010;5(11):e14109. doi: 10.1371/journal.pone.0014109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bais C, Wu X, Yao J, Yang S, Crawford Y, McCutcheon K, et al. PlGF blockade does not inhibit angiogenesis during primary tumor growth. Cell. 2010 Apr 2;141(1):166–77. doi: 10.1016/j.cell.2010.01.033. [DOI] [PubMed] [Google Scholar]
  • 16.Fan F, Wey JS, McCarty MF, Belcheva A, Liu W, Bauer TW, et al. Expression and function of vascular endothelial growth factor receptor-1 on human colorectal cancer cells. Oncogene. 2005 Apr 14;24(16):2647–53. doi: 10.1038/sj.onc.1208246. [DOI] [PubMed] [Google Scholar]
  • 17.Sawano A, Iwai S, Sakurai Y, Ito M, Shitara K, Nakahata T, et al. Flt-1, vascular endothelial growth factor receptor 1, is a novel cell surface marker for the lineage of monocyte-macrophages in humans. Blood. 2001 Feb 1;97(3):785–91. doi: 10.1182/blood.v97.3.785. [DOI] [PubMed] [Google Scholar]
  • 18.Gerber HP, Malik AK, Solar GP, Sherman D, Liang XH, Meng G, et al. VEGF regulates haematopoietic stem cell survival by an internal autocrine loop mechanism. Nature. 2002 Jun 27;417(6892):954–8. doi: 10.1038/nature00821. [DOI] [PubMed] [Google Scholar]
  • 19.Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, et al. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996 Oct;2(10):1096–103. doi: 10.1038/nm1096-1096. [DOI] [PubMed] [Google Scholar]
  • 20.Barleon B, Sozzani S, Zhou D, Weich HA, Mantovani A, Marme D. Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood. 1996 Apr 15;87(8):3336–43. [PubMed] [Google Scholar]
  • 21.Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, et al. Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med. 2004 Feb 12;350(7):672–83. doi: 10.1056/NEJMoa031884. [DOI] [PubMed] [Google Scholar]
  • 22.Ferrara N. Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev. 2004 Aug;25(4):581–611. doi: 10.1210/er.2003-0027. [DOI] [PubMed] [Google Scholar]
  • 23.Ryden L, Linderholm B, Nielsen NH, Emdin S, Jonsson PE, Landberg G. Tumor specific VEGF-A and VEGFR2/KDR protein are co-expressed in breast cancer. Breast Cancer Res Treat. 2003 Dec;82(3):147–54. doi: 10.1023/B:BREA.0000004357.92232.cb. [DOI] [PubMed] [Google Scholar]
  • 24.Giatromanolaki A, Koukourakis MI, Sivridis E, Chlouverakis G, Vourvouhaki E, Turley H, et al. Activated VEGFR2/KDR pathway in tumour cells and tumour associated vessels of colorectal cancer. Eur J Clin Invest. 2007 Nov;37(11):878–86. doi: 10.1111/j.1365-2362.2007.01866.x. [DOI] [PubMed] [Google Scholar]
  • 25.Giatromanolaki A, Koukourakis MI, Turley H, Sivridis E, Harris AL, Gatter KC. Phosphorylated KDR expression in endometrial cancer cells relates to HIF1alpha/VEGF pathway and unfavourable prognosis. Mod Pathol. 2006 May;19(5):701–7. doi: 10.1038/modpathol.3800579. [DOI] [PubMed] [Google Scholar]
  • 26.Ferrara N. Vascular endothelial growth factor as a target for anticancer therapy. Oncologist. 2004;9(Suppl 1):2–10. doi: 10.1634/theoncologist.9-suppl_1-2. [DOI] [PubMed] [Google Scholar]
  • 27.Sun W. Angiogenesis in metastatic colorectal cancer and the benefits of targeted therapy. J Hematol Oncol. 2012;5:63. doi: 10.1186/1756-8722-5-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Van Cutsem E, Tabernero J, Lakomy R, Prenen H, Prausova J, Macarulla T, et al. Addition of Aflibercept to Fluorouracil, Leucovorin, and Irinotecan Improves Survival in a Phase III Randomized Trial in Patients With Metastatic Colorectal Cancer Previously Treated With an Oxaliplatin-Based Regimen. J Clin Oncol. 2012 Sep 4; doi: 10.1200/JCO.2012.42.8201. [DOI] [PubMed] [Google Scholar]
  • 29.Ramlau R, Gorbunova V, Ciuleanu TE, Novello S, Ozguroglu M, Goksel T, et al. Aflibercept and Docetaxel Versus Docetaxel Alone After Platinum Failure in Patients With Advanced or Metastatic Non-Small-Cell Lung Cancer: A Randomized, Controlled Phase III Trial. J Clin Oncol. 2012 Sep 10; doi: 10.1200/JCO.2012.42.6932. [DOI] [PubMed] [Google Scholar]
  • 30.Sanofi and Regeneron Announce Regulatory and Clinical Update for ZALTRAP® (aflibercept) April 5, 2012.
  • 31.Phase 3 Trial of Aflibercept in Metastatic Pancreatic Cancer Discontinued September 11, 2009.
  • 32.Ivy SP, Wick JY, Kaufman BM. An overview of small-molecule inhibitors of VEGFR signaling. Nat Rev Clin Oncol. 2009 Oct;6(10):569–79. doi: 10.1038/nrclinonc.2009.130. [DOI] [PubMed] [Google Scholar]
  • 33•.Witte L, Hicklin DJ, Zhu Z, Pytowski B, Kotanides H, Rockwell P, et al. Monoclonal antibodies targeting the VEGF receptor-2 (Flk1/KDR) as an anti-angiogenic therapeutic strategy. Cancer Metastasis Rev. 1998 Jun;17(2):155–61. doi: 10.1023/a:1006094117427. One of first studies demonstrating effect of VEGFR2 inhibition in vitro. [DOI] [PubMed] [Google Scholar]
  • 34•.Prewett M, Huber J, Li Y, Santiago A, O’Connor W, King K, et al. Antivascular endothelial growth factor receptor (fetal liver kinase 1) monoclonal antibody inhibits tumor angiogenesis and growth of several mouse and human tumors. Cancer Res. 1999 Oct 15;59(20):5209–18. Report tumor growth inhibition of xenograft in vivo by targeting flk1. [PubMed] [Google Scholar]
  • 35•.Lu D, Shen J, Vil MD, Zhang H, Jimenez X, Bohlen P, et al. Tailoring in vitro selection for a picomolar affinity human antibody directed against vascular endothelial growth factor receptor 2 for enhanced neutralizing activity. J Biol Chem. 2003 Oct 31;278(44):43496–507. doi: 10.1074/jbc.M307742200. Details generation of high-affinity, anti-KDR antibody. [DOI] [PubMed] [Google Scholar]
  • 36.Miao HQ, Hu K, Jimenez X, Navarro E, Zhang H, Lu D, et al. Potent neutralization of VEGF biological activities with a fully human antibody Fab fragment directed against VEGF receptor 2. Biochem Biophys Res Commun. 2006 Jun 23;345(1):438–45. doi: 10.1016/j.bbrc.2006.04.119. [DOI] [PubMed] [Google Scholar]
  • 37•.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 Feb 10;28(5):780–7. doi: 10.1200/JCO.2009.23.7537. Largest phase I study demonstrating safety of Ramucirumab in human. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Franklin MC, Navarro EC, Wang Y, Patel S, Singh P, Zhang Y, et al. The structural basis for the function of two anti-VEGF receptor 2 antibodies. Structure. 2011 Aug 10;19(8):1097–107. doi: 10.1016/j.str.2011.01.019. [DOI] [PubMed] [Google Scholar]
  • 39.Chiorean E, S C, Hurwitz H, Savage S, Cohen R, Schwartz J, Wang G, Fox F, Rowinsky E, Youssoufian H. Phase I dose-escalation study of the anti-VEGFR-2 recombinant human IgG1 MAb IMC-1121B administered every other week (q2w) or every 3 weeks (q3w) in patients (pts) with advanced cancers. 2007 [Google Scholar]
  • 40.Spratlin J. Ramucirumab (IMC-1121B): Monoclonal antibody inhibition of vascular endothelial growth factor receptor-2. Curr Oncol Rep. 2011 Apr;13(2):97–102. doi: 10.1007/s11912-010-0149-5. [DOI] [PubMed] [Google Scholar]
  • 41.Zhu AX, Finn RS, Mulcahy MF, Gurtler JS, Sun W, Schwartz JD, et al. A phase II study of ramucirumab as first-line monotherapy in patients (pts) with advanced hepatocellular carcinoma (HCC). ASCO Meeting Abstracts; 2010 June 14; p. 4083. [Google Scholar]
  • 42.Camidge DR, Ballas MS, Dubey S, Haigentz M, Rosen PJ, Spicer JF, et al. A phase II, open-label study of ramucirumab (IMC-1121B), an IgG1 fully human monoclonal antibody (MAb) targeting VEGFR-2, in combination with paclitaxel and carboplatin as first-line therapy in patients (pts) with stage IIIb/IV non-small cell lung cancer (NSCLC). ASCO Meeting Abstracts; 2010 June 14; p. 7588. [Google Scholar]
  • 43.Carvajal RD, Wong MK, Thompson JA, Gordon MS, Lewis KD, Pavlick AC, et al. A phase II randomized study of ramucirumab (IMC-1121B) with or without dacarbazine (DTIC) in patients (pts) with metastatic melanoma (MM). ASCO Meeting Abstracts; 2010 June 14; p. 8519. [Google Scholar]
  • 44.Penson RT, Moore KN, Fleming GF, Braly PS, Schimp VL, Nguyen H, et al. A phase II, open-label, multicenter study of IMC-1121B (ramucirumab; RAM) monotherapy in the treatment of persistent or recurrent epithelial ovarian (EOC), fallopian tube (FTC), or primary peritoneal (PPC) carcinoma (CP12-0711/ NCT00721162). ASCO Meeting Abstracts; 2012 May 30; p. 5012. [Google Scholar]
  • 45.Hussain M, Rathkopf DE, Liu G, Armstrong AJ, Kelly WK, Ferrari AC, et al. A phase II randomized study of cixutumumab (IMC-A12: CIX) or ramucirumab (IMC-1121B: RAM) plus mitoxantrone (M) and prednisone (P) in patients (pts) with metastatic castrate-resistant prostate cancer (mCRPC) following disease progression (PD) on docetaxel (DCT) therapy. ASCO Meeting Abstracts; 2012 February 2; p. 97. [Google Scholar]
  • 46.Garcia-Carbonero R, Rivera F, Maurel J, Ayoub J-PM, Moore MJ, Cervantes-Ruiperez A, et al. A phase II, open-label study evaluating the safety and efficacy of ramucirumab combined with mFOLFOX-6 as first-line therapy in patients (pts) with metastatic colorectal cancer (mCRC): CP12-0709/ NCT00862784. ASCO Meeting Abstracts; 2012 January 30; p. 533. [Google Scholar]
  • 47.Vahdat LT, Miller K, Sparano JA, Youssoufian H, Schwartz JD, Nanda S, et al. Randomized phase II study of capecitabine with or without ramucirumab (IMC-1121B) or IMC-18F1 in patients with unresectable, locally advanced or metastatic breast cancer (mBC) previously treated with anthracycline and taxane therapy (CP20-0903/ NCT01234402). ASCO Meeting Abstracts; 2011 June 9; TPS151. [Google Scholar]
  • 48.Petrylak DP, Chi KN, Vogelzang NJ, Sonpavde G, Rutstein MD, Schwartz JD, et al. Randomized phase II study of docetaxel with or without ramucirumab (IMC-1121B) or icrucumab (IMC-18F1) in patients with urothelial transitional cell carcinoma (TCC) following progression on first-line platinum-based therapy. ASCO Meeting Abstracts; 2012 May 30; TPS4675. [Google Scholar]
  • 49•.Fuchs CS, Tomasek J, Cho JY, Dumitru F, Passalacqua R, Goswami C, et al. REGARD: A phase III, randomized, double-blinded trial of ramucirumab and best supportive care (BSC) versus placebo and BSC in the treatment of metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma following disease progression on first-line platinum- and/or fluoropyrimidine-containing combination therapy. ASCO Meeting Abstracts; 2013 January 30; LBA5, First abstract showing efficacy of Ramucirumab in metastatic gastric cancer. [Google Scholar]
  • 50•.Fuchs CS, T J, Cho JY, Filip D, Passalacqua R, Goswami C, Safran H, Dos Santos LV, Aprile G, Ferry D, Melichar B, Tehfe M, Topuzov E, Tabernero J, Zalcberg R, Chau I, Koshiji M, Hsu Y, Schwartz JD, Ajani J. REGARD: A phase III, randomized, double-blind trial oframucirumab and best supportive care (BSC) versus placebo and BSC in the treatment of metastatic gastric orgastroesophageal junction (GEJ) adenocarcinoma following disease progression on first-line platinum- and/or fluoropyrimidine-containing combination therapy. Annual Meeting 2013; Washington DC. American Association for Cancer Research; 2013. Full results of REGARD trial presented. [Google Scholar]
  • 51.Yi JH, Lee J, Park SH, Park JO, Yim DS, Park YS, et al. Randomised phase II trial of docetaxel and sunitinib in patients with metastatic gastric cancer who were previously treated with fluoropyrimidine and platinum. Br J Cancer. 2012 Apr 24;106(9):1469–74. doi: 10.1038/bjc.2012.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sun W, Powell M, O’Dwyer PJ, Catalano P, Ansari RH, Benson AB., 3rd Phase II study of sorafenib in combination with docetaxel and cisplatin in the treatment of metastatic or advanced gastric and gastroesophageal junction adenocarcinoma: ECOG 5203. J Clin Oncol. 2010 Jun 20;28(18):2947–51. doi: 10.1200/JCO.2009.27.7988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.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 Oct 20;29(30):3968–76. doi: 10.1200/JCO.2011.36.2236. [DOI] [PubMed] [Google Scholar]
  • 54.Thuss-Patience PC, Kretzschmar A, Bichev D, Deist T, Hinke A, Breithaupt K, et al. Survival advantage for irinotecan versus best supportive care as second-line chemotherapy in gastric cancer--a randomised phase III study of the Arbeitsgemeinschaft Internistische Onkologie (AIO) Eur J Cancer. 2011 Oct;47(15):2306–14. doi: 10.1016/j.ejca.2011.06.002. [DOI] [PubMed] [Google Scholar]
  • 55.Kang JH, Lee SI, Lim do H, Park KW, Oh SY, Kwon HC, et al. Salvage chemotherapy for pretreated gastric cancer: a randomized phase III trial comparing chemotherapy plus best supportive care with best supportive care alone. J Clin Oncol. 2012 May 1;30(13):1513–8. doi: 10.1200/JCO.2011.39.4585. [DOI] [PubMed] [Google Scholar]
  • 56.Zhu AX, Chau I, Blanc J-F, Okusaka T, Rojas M, Yang L, et al. A multicenter, randomized, double-blind, phase III study of ramucirumab (IMC-1121B; RAM) and best supportive care (BSC) versus placebo (PBO) and BSC as second-line treatment in patients (pts) with hepatocellular carcinoma (HCC) following first-line therapy with sorafenib (SOR). ASCO Meeting Abstracts; 2012 May 30; TPS4146. [Google Scholar]
  • 57. [2013 April 10];Full Prescribing Information: Stivarga. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/203085lbl.pdf.
  • 58. [2013 April 10];Full Prescribing Information: Avastin. Available from: http://www accessdata.fda.gov/drugsatfda_docs/label/2013/125085s267lbl.pdf.
  • 59.Full Prescribing Information: Zaltrap. [2013 April 10]; Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/125418s000lbl.pdf.
  • 60.Bottsford-Miller JN, Coleman RL, Sood AK. Resistance and escape from antiangiogenesis therapy: clinical implications and future strategies. J Clin Oncol. 2012 Nov 10;30(32):4026–34. doi: 10.1200/JCO.2012.41.9242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008 Aug;8(8):592–603. doi: 10.1038/nrc2442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Leppanen VM, Jeltsch M, Anisimov A, Tvorogov D, Aho K, Kalkkinen N, et al. Structural determinants of vascular endothelial growth factor-D receptor binding and specificity. Blood. 2011 Feb 3;117(5):1507–15. doi: 10.1182/blood-2010-08-301549. [DOI] [PubMed] [Google Scholar]
  • 63.Lieu CH, Tran HT, Jiang Z, Mao M, Overman MJ, Eng C, et al. The association of alternate VEGF ligands with resistance to anti-VEGF therapy in metastatic colorectal cancer. ASCO Meeting Abstracts; 2011 June 9; p. 3533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64•.Nixon AB, Pang H, Starr M, Hollis D, Friedman PN, Bertagnolli MM, et al. Prognostic and predictive blood-based biomarkers in patients with advanced pancreatic cancer: Results from CALGB 80303. Journal of Clinical Oncology. 2011;29(suppl) doi: 10.1158/1078-0432.CCR-13-0926. abstr 10508. Demonstrates predictive value of VEGFD for bevacizumab in pancreatic cancer patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65•.Weickhardt AJ, W D, Lee C, Simes J, Murone C, Wilson K, Cummins M, Asadi K, Price TJ, Mariadason J, Tebbutt NC. Vascular endothelial growth factors (VEGF) and VEGF receptor expression as predictive biomarkers for benefit with bevacizumab in metastatic colorectal cancer (mCRC): Analysis of the phase III MAX study. J Clin Oncol. 2011;29(suppl) abstr 3531. Demonstrate predictive value of VEGFD for bevacizumab in colorectal cancer patients. [Google Scholar]
  • 66.Gabrilovich D, Ishida T, Oyama T, Ran S, Kravtsov V, Nadaf S, et al. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood. 1998 Dec 1;92(11):4150–66. [PubMed] [Google Scholar]
  • 67.Osada T, Chong G, Tansik R, Hong T, Spector N, Kumar R, et al. The effect of anti-VEGF therapy on immature myeloid cell and dendritic cells in cancer patients. Cancer Immunol Immunother. 2008 Aug;57(8):1115–24. doi: 10.1007/s00262-007-0441-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wu Y, Hooper AT, Zhong Z, Witte L, Bohlen P, Rafii S, et al. The vascular endothelial growth factor receptor (VEGFR-1) supports growth and survival of human breast carcinoma. Int J Cancer. 2006 Oct 1;119(7):1519–29. doi: 10.1002/ijc.21865. [DOI] [PubMed] [Google Scholar]

RESOURCES