Next Article in Journal
Gestational Diabetes Mellitus: The Dual Risk of Small and Large for Gestational Age: A Narrative Review
Previous Article in Journal / Special Issue
Current and Emerging Therapeutic Strategies for Limited- and Extensive-Stage Small-Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Combining Immune Checkpoint Inhibitors and Anti-Angiogenesis Approaches: Treatment of Advanced Non-Small Cell Lung Cancer

1
Boonshoft School of Medicine, Dayton, OH 45435, USA
2
Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Dayton, OH 45435, USA
*
Author to whom correspondence should be addressed.
Med. Sci. 2025, 13(3), 143; https://doi.org/10.3390/medsci13030143
Submission received: 23 June 2025 / Revised: 1 August 2025 / Accepted: 12 August 2025 / Published: 19 August 2025
(This article belongs to the Special Issue Feature Papers in Section Cancer and Cancer-Related Diseases)

Abstract

Combining immune checkpoint inhibitors (ICIs) and anti-angiogenic pharmacologic agents is an encouraging therapeutic approach in the treatment of non-small cell lung cancer (NSCLC). Currently, the only FDA-approved therapy combining an immune checkpoint inhibitor and a vascular endothelial growth factor (VEGF) inhibitor is atezolizumab, bevacizumab, and chemotherapy in first-line metastatic NSCLC patients. However, the combination of nivolumab, a programmed death-1 (PD-1) inhibitor, and bevacizumab has also shown encouraging results in patients with NSCLC with minimal adverse effects, respectively. This communication aims to highlight the efficacy of nivolumab and bevacizumab in NSCLC patients without sensitizing mutations in epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), or ROS proto-oncogene 1 (ROS1). In addition, the combination of nivolumab/atezolizumab and bevacizumab with other therapeutic agents is also discussed. We also underscore the adverse effects and limitations of such combinations in NSCLC patients. Future studies should focus on large-scale trials and biomarker identification to establish the benefits of these combination therapies in NSCLC patients.

1. Introduction

Lung cancer is one of the leading causes of cancer-related mortality worldwide and is the major cause of cancer deaths in the United States [1,2,3]. Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, accounting for approximately 85% of all lung cancer cases [4,5]. Despite recent advancements in treatment modalities, the prognosis for patients with advanced or metastatic NSCLC without driver mutations remains poor [6,7]. Effective treatment options remain limited, especially for patients with resistance to standard therapies, but immunotherapy has changed the treatment of NSCLC, first in second-line treatment, then in first-line treatment, and now also in neoadjuvant and perioperative treatment [6,8,9]. This study is timely due to the lack of clinical trial data over the past 5 years discussing the combination of nivolumab and bevacizumab in NSCLC patients without sensitizing gene mutations. There is increasing clinical interest in identifying therapies to modulate tumor responses, but further studies of efficacy are warranted. The advent of immune checkpoint inhibitors (ICIs) targeting programmed death-1 (PD-1)/PD-ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) have revolutionized cancer therapy due to their ability to utilize the body’s own immune system to identify and destroy cancer cells [10,11,12,13]. However, ICIs face challenges due to immune evasion mechanisms within the tumor microenvironment (TME) [14,15]. ICIs foster a strong immune response, which enables the immune system, particularly T cells within TME, to better recognize and attack tumor cells. However, resistance to ICIs poses significant challenges to effective treatment options [14,15]. Given the critical role of angiogenesis in tumor survival and its impact on tumor progression, it has become a promising focus of research. Current efforts aim to overcome resistance to immunotherapy in lung cancer by targeting angiogenic pathways and exploring therapeutic strategies that combine immune checkpoint inhibitors (ICIs) with anti-angiogenic agents to enhance treatment efficacy and improve patient outcomes [16,17,18].
Notably, ICIs such as nivolumab, a monoclonal antibody targeting the PD-1 receptor, has demonstrated significant efficacy in NSCLC by disrupting the interaction of the PD-1 receptor with its ligands PD-L1 and PD-L2, thereby inhibiting the cellular immune response [19,20]. In addition, anti-angiogenic agents like bevacizumab, a targeted vascular endothelial growth factor (VEGF) inhibitor, have shown the ability to disrupt tumor vasculature, thereby inhibiting tumor growth and metastasis [21,22,23]. The idea for combining nivolumab with bevacizumab lies in their complementary mechanisms of action: while nivolumab stimulates T-cell-mediated immune responses, bevacizumab reduces angiogenesis, which can modify the TME to enhance immune cell infiltration and function [19,21,22,23]. This combination therapy has the potential to overcome resistance mechanisms that limit the efficacy of monotherapies, offering a more comprehensive approach to treating NSCLC. Clinical trials have provided encouraging results, showing improved PFS and OS in patients treated with the combination of nivolumab and bevacizumab, particularly when used alongside platinum-based chemotherapy [24,25,26,27]. The goal of this communication is to discuss the efficacy of targeting immune checkpoints and angiogenesis in patients without having sensitizing mutations in epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), or ROS proto-oncogene 1 (ROS1) oncogenes, and to highlight the exploration of combination therapy for NSCLC treatment without utilizing predictive biomarker responses.

2. Mechanisms of Action of Nivolumab and Bevacizumab

Nivolumab (OPDIVO) is an FDA-approved drug available as a single-use injection for a variety of human malignancies, including NSCLC, small cell lung cancer (SCLC), pleural mesothelioma, melanoma, squamous cell carcinoma of the head and neck, renal cell carcinoma, urothelial carcinoma, hepatocellular carcinoma, colorectal cancer, Hodgkin lymphoma, and esophageal squamous cell carcinoma [19,28]. Nivolumab is a human IgG4 monoclonal antibody checkpoint inhibitor that binds to programmed death-1 receptors (PD-1) expressed on activated T-cells. When PD-1 binds to its ligands, PD-L1 and PD-L2, which are expressed on antigen presenting cells (APCs) and other cells, inhibitory signals are sent to T-cells. This dampens the immune response, helping to maintain tolerance and prevent tissue damage from excessive immune activity [19]. However, abnormal PD-L1 expression by tumor cells utilize this pathway to evade proper immune detection [19]. When PD-L1 on tumor cells binds to PD-1 on T-cells, the lymphocytes become deactivated and allow the tumor cells to evade immune recognition and continue proliferation unchecked. Nivolumab blocks the interaction between PD-1 and PD-L1/PD-L2 to prevent the inhibitory signal from being transmitted, allowing T-cells to remain active and recognize and destroy tumor cells [19,29,30].
Bevacizumab (AVASTIN) was first approved by the FDA in 2004 as an intravenous (IV) infusion and is currently approved for metastatic colorectal cancer, non-squamous NSCLC, glioblastoma, metastatic renal cell carcinoma, and persistent, recurrent, or metastatic carcinoma of the cervix [31]. Bevacizumab is a recombinant humanized monoclonal IgG1 antibody that binds to VEGF to inhibit its binding to the cell surface VEGF receptors for both the tumor cells as well as the local healthy cells [31]. This creates two different mechanisms by which bevacizumab can combat cancer cells. By blocking VEGF from binding to healthy endothelial cells, it can effectively block the signaling for angiogenesis and disrupt the creation of blood vessels needed by the tumor cells [17,21,22,32,33]. When VEGF is blocked from binding to the receptors directly on the tumor cells, it can inhibit the signaling for cell proliferation and survival. VEGF is also an important immunomodulator of TME. The mechanism of actions of nivolumab and bevacizumab is depicted in Figure 1.
When used in combination, nivolumab and bevacizumab offer complimentary mechanisms. Despite this synergy, resistance to the combination may still arise. VEGF inhibitors can lead to hypoxia driven upregulation of other pro-angiogenic pathways. Similarly, not all tumors respond to PD-1 blockage due to antigen presentation defects.

3. Mechanism of Resistance and Adverse Reactions of Nivolumab and Bevacizumab

Patients treated with immune checkpoint therapy such as nivolumab can acquire resistance to the drug through mechanisms that are not very well understood [27]. Many theorize that resistance could be multifactorial, including ideas such as the TME including various factors and cell types such as regulatory T cells (Tregs), myeloid-derived suppressive cells (MDSCs), tumor-associated macrophages (TAMs), and immature dendritic cells (imDC) [18]. Low or variable PD-L1 expression in NSCLC tumors and other associated mechanisms might not respond well to nivolumab [27]. Certain mutations in genes such as rearranged during transfection (RET) or human epidermal growth factor receptor 2 (HER2) can also reduce the expression of PD-L1, thus reducing the effectiveness of nivolumab [27]. Additionally, low tumor mutation burden (TMB) can increase the likelihood of nivolumab and other PD-1 inhibitors being an ineffective treatment option because these tumors are unable to elicit a strong immune response to PD-1 inhibiting drugs [10,34]. Other mechanisms of resistance involve insufficient antigen recognition by T-cells and other downstream effects of issues in the T-cell activation process [35]. Additional factors that affect the resistance of ICIs such as nivolumab include EGFR mutations and ALK alterations [36]. Moreover, associated adverse reactions are other potential challenges to effective treatments. The FDA cites several adverse reactions to nivolumab as a single agent as well as in combination with ipilimumab, platinum chemotherapy, cabozantinib and with fluoropyrimidine [28]. For example, nivolumab can cause severe infusion-related reactions, which have been reported in <1.0% of patients in clinical trials [28]. Fatigue, musculoskeletal pain, diarrhea, nausea, rash, pruritus, asthenia, cough, dyspnea, constipation, decreased appetite, back pain, upper respiratory tract infection, arthralgia, pyrexia, headache, abdominal pain, vomiting, and urinary tract infection are other adverse effects of nivolumab [28].
While bevacizumab impacts tumor proliferation in multiple ways, patients receiving bevacizumab have shown acquired resistance to the treatment [32]. The specific mechanisms of resistance of bevacizumab are still being studied; however, there are several factors that are thought to play a role in the resistance [32,33]. First, while VEGF is a powerful angiogenic signaling molecule, it is not the only signal for angiogenesis, and it is thought that the tumor cells can upregulate other angiogenic signaling pathways, such as hypoxia-inducible factor-1 alpha (HIF-1α) in the presence of bevacizumab [32]. Second, the TME seems to broadly impact the efficacy of chemotherapeutic agents and bevacizumab. It seems hypoxia is the most significant factor in the TME, which is consequently increased by blocking angiogenesis with a VEGF inhibitor [32,35,37]. In fact, it is documented that the hypoxia induced by an angiogenesis inhibitor has the potential to worsen tumor invasiveness and metastasis [37]. Notably, there are several implications of hypoxia in the TME that can lead to tumor survival, but among them is the hypoxia’s recruitment of myeloid cells into the TME that ultimately facilitate an immunosuppressive microenvironment and a weakened antitumor response [32]. Moreover, induction of hypoxic condition mediated through VEGF inhibitors reduces the recruitment of suppressive cells into the TME to improve the efficacy of immunotherapies [18,27]. Additionally, adverse reactions to bevacizumab are also potential challenges to effective treatment. Bevacizumab is also associated with several adverse reactions that occurred in greater than 10% of patients taking the medication. These adverse reactions include epistaxis, headache, hypertension, rhinitis, proteinuria, taste alteration, dry skin, rectal hemorrhage, lacrimation disorder, back pain, and exfoliative dermatitis [31]. According to the FDA, bevacizumab was discontinued in 8.4–21% of patients across all studies because of these reactions. Of note, the maximum dose tested in humans was 20 mg/kg IV, and nine of the sixteen patients developed a headache, three of which experienced severe headaches [31].

4. Studies of Nivolumab and Bevacizumab Combination with or Without Other Agents

A focused literature search was conducted using PubMed for articles published between 2020 and 2024. Keywords included ‘non-small cell lung cancer,’ ‘nivolumab,’ ‘bevacizumab,’ ‘immune checkpoint inhibitors,’ and ‘anti-angiogenesis.’ Only English-language articles and clinical trials involving human subjects were included, yielding the four studies discussed below. The two drugs, nivolumab and bevacizumab, have been studied in clinical trials in addition to therapies such as platinum chemotherapy, cell cycle inhibitor chemotherapy, and DNA synthesis inhibitor chemotherapy [24,25,26,38]. There is a gap in our knowledge of how nivolumab and bevacizumab combination without additional chemotherapy interact as a potential therapy for treatment in NSCLC. The summary of clinical studies with a combination of nivolumab and bevacizumab is given in Table 1.
One clinical trial assessed the approach of combining nivolumab or placebo with bevacizumab plus platinum-based chemotherapy for treatment of NSCLC patients without driver mutations in ALK, EGFR, or ROS1, evaluated the safety and effectiveness in the treatment plan [24]. The 550 patients were randomized. At the time of the analysis (minimum follow-up: 19.4 months), the median overall survival (OS) was longer in the nivolumab arm than in the placebo arm (30.8 vs. 24.7 months; hazard ratio 0.74; 95% confidence interval 0.58–0.94). The 12-month OS rates were 81.3% vs. 76.3% in the nivolumab vs. placebo arms, respectively. The respective 18-month OS rates were 69.0% vs. 61.9%. This treatment regimen was identical to their previous clinical trial and concluded that nivolumab in combination with platinum chemotherapy and bevacizumab demonstrated a longer OS compared to the placebo combination [24]. This study did not mention adverse events in their conclusion.
In an additional study, nivolumab was assessed with carboplatin, paclitaxel, and bevacizumab in NSCLC patients with stage IIIB/IV or recurrent NSCLC without sensitizing EGFR, ALK, or ROS1 alterations [25]. This study initially used carboplatin, paclitaxel, and bevacizumab every 3 weeks for up to six cycles and had maintenance therapy using nivolumab/placebo with bevacizumab [25]. This study included a cohort of 550 patients, 273 received the nivolumab and 275 received placebo combinations, respectively. With a median follow up of 13.7 months, the Independent Regulatory Review (IRRC)-assessed that the median progression free survival (PFS) was significantly longer in the nivolumab arm than in the placebo arm (12.1 versus 8.1 months; hazard ratio 0.56; 96.4% confidence interval 0.43–0.71). The PFS benefit was observed across all patients with any PD-L1 expression levels, including PD-L1-negative patients. This study concluded that nivolumab/bevacizumab with cytotoxic chemotherapy should be considered as it demonstrated significant improvement in PFS [25]. The incidence of treatment related adverse events was comparable between the control and treatment groups. Treatment related events leading to death were observed in five and four patients in the treatment and placebo group, respectively [25].
An additional clinical trial assessed nivolumab efficacy in different combinations of chemotherapeutic agents for NSCLC without targetable oncogenes, respectively [26]. The patients were divided into four treatment arms. Arm A includes four cycles of cisplatin and gemcitabine; arm B includes four cycles of cisplatin and pemetrexed followed by pemetrexed maintenance therapy; arm C includes four to six cycles of carboplatin, paclitaxel, and bevacizumab followed by bevacizumab; and arm D includes docetaxel. In this trial, minimum follow-up period was 57.9 months. Median PFS was 6.3 (0.7+–47.8), 11.8 (1.4–65.1+), 40.7 (5.3–60.8+), and 3.2 (1.9–10.9) months, and 5-year PFS was observed in 0/6, 1/6, 1/6, and 0/6 patients in arms A, B, C, and D, respectively. Median OS was 13.2 (11.0–55.4), 28.5 (14.6–66.2+), not reached (24.2–67.4+), and 12.5 (9.8–16.9) months, and the number of patients surviving 5 years were 0/6, 1/6, 4/6, and 0/6 in arms A, B, C, and D, respectively. This trial concluded that the nivolumab/carboplatin/paclitaxel/bevacizumab therapy combination showed both tolerability and 5-year PFS and OS [26]. No unexpected severe adverse events or treatment-related deaths occurred in this trial [26].
A systematic review assessed checkpoint inhibitor combinations, including pembrolizumab, atezolizumab, and atezolizumab/bevacizumab for NSCLC treatment. This review showed significantly improved OS compared with controls in patients with advanced NSCLC without EGFR/ALK mutations [38]. Two trials reported outcomes for squamous NSCLC, with pembrolizumab–chemotherapy reporting significantly improved (OS) compared with chemotherapy. Of note, the combination of nivolumab–ipilimumab failed to improve OS [38]. Outcomes for atezolizumab–bevacizumab–chemotherapy in EGFR+/ALK+ patients are promising and require further exploration. The results of this review indicated that pembrolizumab (PD-1 inhibitor) improved OS, and the atezolizumab/bevacizumab combination therapy for EGFR+/ALK+ patients requires additional studies [38].
While these studies collectively demonstrate the potential of combining ICIs and anti-angiogenesis agents to benefit patients with NSCLC, there are important limitations and discrepancies. Variations in trial design, chemotherapy regimens, follow-up periods, and reporting of adverse events limit direct comparison. This highlights the need for standardized study designs and biomarker-driven strategies in future studies.

5. Potential of Other ICIs and Anti-Angiogenic Therapies for NSCLC

In addition to the combination of nivolumab and bevacizumab, several other ICIs have been studied in combination with anti-angiogenic therapies for the treatment of NSCLC [18,39]. The other considerable therapies are listed in Table 2.
Atezolizumab, an anti-PD-L1 antibody, has been evaluated in combination with bevacizumab in a phase III clinical trial [18]. This study demonstrated that the combination, along with chemotherapy, improved PFS and OS in patients with metastatic non-squamous NSCLC [18]. The median PFS of the group in combination with atezolizumab, carboplatin, paclitaxel, and bevacizumab was 8.3 months, and the carboplatin, paclitaxel, and bevacizumab group PFS was 6.8 months (HR: 0.59). The median OS was 19.2 months for the first group, and 14.7 months for the second 31 group (HR: 0.78) [18]. The anti-angiogenic effects of bevacizumab are thought to enhance the immune response elicited by atezolizumab. The incidence of treatment-related adverse events was 25.4% for the atezolizumab, carboplatin, paclitaxel, and bevacizumab group and 19.3% for the bevacizumab, carboplatin, and paclitaxel group. However, 77.4% of atezolizumab, bevacizumab, carboplatin, and paclitaxel (ABCP) patients had grade 1–2 AEs [18].
Pembrolizumab has been combined with lenvatinib, a tyrosine kinase inhibitor (TKI) that includes anti-angiogenic properties such as targeting VEGF and fibroblast growth factor (FGF) [39]. This combination has shown potential in enhancing anti-tumor activity by modulating the TME [39]. In addition, preclinical studies suggest that lenvatinib can decrease TAMs and Tregs, thereby improving the efficacy of ICIs [39,40].
Pembrolizumab has also been combined with ramucirumab, an anti-VEGFR-2 antibody [40]. An open-label phase 1a/b trial found median PFS at 9.3 months, while 12-month and 18-month PFS were each 45%. Median OS was not reached, but the 12-month and 18-month OS rates were 73% and 64%, respectively [40]. The rationale is that inhibiting VEGFR-2 can normalize tumor vasculature and reduce immunosuppressive cells, thereby potentiating the effects of pembrolizumab [40]. Adverse effects associated with this therapy were fatigue and myocardial infarction in 7% of 31 patients [18,40].

6. Conclusions and Future Perspectives

NSCLC has a variety of treatment options for patients lacking EGFR, ALK, or ROS1 mutations. The data demonstrated that nivolumab in combination with bevacizumab and platinum-based chemotherapy provide promising results in improving PFS and OS in patients with NSCLC. While few clinical trials demonstrated the beneficial effects of these immunotherapies, further research is needed to assess the efficacy of combination immunotherapy in patients lacking gene mutations. In addition, while biomarkers aid in the diagnosis of disease pathology, identifying biomarkers that predict response to combination therapy remains a challenge. Moreover, lack of accuracy in biomarkers can lead to unnecessary pharmacological interventions and potential adverse effects which should be monitored carefully in patients receiving treatment. While biomarkers such as PD-L1 expression are useful in predicting nivolumab efficacy, they are not reliable predictors of adverse events. Although nivolumab generally has a favorable safety profile, further research is needed to identify biomarkers that can guide both therapeutic response and toxicity risk. Bevacizumab has been reported to have adverse reactions in greater than 10% of patients. Taken together, nivolumab and bevacizumab should be evaluated with careful consideration with other therapeutic agents for the treatment of NSCLC. Additionally, current clinical data on ICIs and anti-angiogenesis combinations in NSCLC are currently limited by small sample sizes and short follow-up periods and should aim to incorporate more diverse patient populations. Emphasis should be placed on longitudinal sampling to monitor changes in the TME and immune system functionality. Future research should focus on developing more comprehensive biomarker panels that combine immune markers, angiogenesis markers, and characteristics of the TME. Gaining a better understanding of resistance mechanisms could help inform the design of combination therapies and improve how treatments are timed to extend patient response.

Author Contributions

Conceptualization, T.B. and R.P.S.; BioRender software, A.T.; validation, T.B., A.T. and R.P.S.; formal analysis, T.B.; data curation, T.B.; writing—original draft preparation, T.B.; writing—review and editing, T.B., A.T. and R.P.S.; visualization, A.T.; supervision, R.P.S.; project administration, R.P.S. All authors have read and agreed to the published version of the manuscript.

Funding

The financial support from the NIH R21 Grant ES033806 (to R.P.S.) is greatly appreciated.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

The authors would like to acknowledge BioRender software (https://www.biorender.com/) for generating the figure.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

NSCLCNon-small cell lung cancer
ICIsImmune checkpoint inhibitors
PFSProgression free survival
OSOverall survival
EGFREpidermal growth factor receptor
ALKAnaplastic lymphoma kinase
ROS1ROS proto-oncogene 1
PD-1Programmed death-1
PD-L1PD-ligand 1
CTLA-4cytotoxic T lymphocyte antigen 4
TMETumor microenvironment
VEGFVascular endothelial growth factor
APCAntigen presenting cells
TregsRegulatory T cells
MDSCsMyeloid-derived suppressive cells
TAMsTumor-associated macrophages
imDCImmature dendritic cells
RETRearranged during transfection
TMBTumor mutation burden
HIF-1αHypoxia-inducible factor-1 alpha
IRRCIndependent Regulatory Review
FGFFibroblast growth factor
TKITyrosine kinase inhibitor

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  2. Lahiri, A.; Maji, A.; Potdar, P.D.; Singh, N.; Parikh, P.; Bisht, B.; Mukherjee, A.; Paul, M.K. Lung cancer immunotherapy: Progress, pitfalls, and promises. Mol. Cancer 2023, 22, 40. [Google Scholar] [CrossRef]
  3. Fois, S.S.; Paliogiannis, P.; Zinellu, A.; Fois, A.G.; Cossu, A.; Palmieri, G. Molecular Epidemiology of the Main Druggable Genetic Alterations in Non-Small Cell Lung Cancer. Int. J. Mol. Sci. 2021, 22, 612. [Google Scholar] [CrossRef] [PubMed]
  4. Basumallik, N.; Agarwal, M. Small Cell Lung Cancer. StatPearls, 10 July 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK482458/ (accessed on 15 March 2025).
  5. Zhu, J.; Li, R.; Tiselius, E.; Roudi, R.; Teghararian, O.; Suo, C.; Song, H. Immunotherapy (excluding checkpoint inhibitors) for stage I to III non-small cell lung cancer treated with surgery or radiotherapy with curative intent. Cochrane Database Syst. Rev. 2017, 12, CD011300. [Google Scholar] [CrossRef]
  6. Miao, D.; Zhao, J.; Han, Y.; Zhou, J.; Li, X.; Zhang, T.; Li, W.; Xia, Y. Management of locally advanced non-small cell lung cancer: State of the art and future directions. Cancer Commun. 2024, 44, 23–46. [Google Scholar] [CrossRef] [PubMed]
  7. Dafni, U.; Tsourti, Z.; Vervita, K.; Peters, S. Immune checkpoint inhibitors, alone or in combination with chemotherapy, as first-line treatment for advanced non-small cell lung cancer. A systematic review and network meta-analysis. Lung Cancer 2019, 134, 127–140. [Google Scholar] [CrossRef] [PubMed]
  8. Muthusamy, B.; Patil, P.D.; Pennell, N.A. Perioperative Systemic Therapy for Resectable Non-Small Cell Lung Cancer. J. Natl. Compr. Cancer Netw. 2022, 20, 953–961. [Google Scholar] [CrossRef]
  9. Uprety, D.; West, H.J. Perioperative Therapy for Resectable Non–Small-Cell Lung Cancer: Weighing Options for the Present and Future. JCO Oncol. Pract. 2023, 19, 403–409. [Google Scholar] [CrossRef]
  10. Ricciuti, B.; Wang, X.; Alessi, J.V.; Rizvi, H.; Mahadevan, N.R.; Li, Y.Y.; Polio, A.; Lindsay, J.; Umeton, R.; Sinha, R.; et al. Association of High Tumor Mutation Burden in Non–Small Cell Lung Cancers with Increased Immune Infiltration and Improved Clinical Outcomes of PD-L1 Blockade Across PD-L1 Expression Levels. JAMA Oncol. 2022, 8, 1160–1168. [Google Scholar] [CrossRef]
  11. Wu, K.; Yi, M.; Qin, S.; Chu, Q.; Zheng, X.; Wu, K. The efficacy and safety of combination of PD-1 and CTLA-4 inhibitors: A meta-analysis. Exp. Hematol. Oncol. 2019, 8, 26. [Google Scholar] [CrossRef]
  12. Tanvetyanon, T.; Gray, J.E.; Antonia, S.J. PD-1 checkpoint blockade alone or combined PD-1 and CTLA-4 blockade as immunotherapy for lung cancer? Expert Opin. Biol. Ther. 2017, 17, 305–312. [Google Scholar] [CrossRef]
  13. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef]
  14. Cheng, W.; Kang, K.; Zhao, A.; Wu, Y. Dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 in lung cancer. J. Hematol. Oncol. 2024, 17, 54. [Google Scholar] [CrossRef] [PubMed]
  15. Xia, L.; Liu, Y.; Wang, Y. PD-1/PD-L1 Blockade Therapy in Advanced Non-Small-Cell Lung Cancer: Current Status and Future Directions. Oncologist 2019, 24 (Suppl. 1), S31–S41. [Google Scholar] [CrossRef]
  16. Wang, J.; Peng, W.; Jiang, M.; Wu, L. Research Progress of Anti-angiogenic Agents Combined with Immunotherapy in Patients with Advanced Non-small Cell Lung Cancer. Zhongguo Fei Ai Za Zhi. 2021, 24, 196–203. [Google Scholar] [CrossRef]
  17. Xu, Z.; Li, J. Review on the combination strategy of anti-angiogenic agents and other anti-tumor agents in advanced non-small cell lung cancer. Chin. J. Lung Cancer 2021, 24, 357–364. [Google Scholar] [CrossRef]
  18. Ren, S.; Xiong, X.; You, H.; Shen, J.; Zhou, P. The Combination of Immune Checkpoint Blockade and Angiogenesis Inhibitors in the Treatment of Advanced Non-Small Cell Lung Cancer. Front. Immunol. 2021, 12, 689132. [Google Scholar] [CrossRef] [PubMed]
  19. Rendon, A.; Rayi, A. Nivolumab. StatPearls, 28 February 2024. Available online: https://www.ncbi.nlm.nih.gov/books/NBK567801/ (accessed on 2 January 2025).
  20. Guo, L.; Zhang, H.; Chen, B. Nivolumab as Programmed Death-1 (PD-1) Inhibitor for Targeted Immunotherapy in Tumor. J. Cancer 2017, 8, 410–416. [Google Scholar] [CrossRef] [PubMed]
  21. Garcia, J.; Hurwitz, H.I.; Sandler, A.B.; Miles, D.; Coleman, R.L.; Deurloo, R.; Chinot, O.L. Bevacizumab (Avastin®) in cancer treatment: A review of 15 years of clinical experience and future outlook. Cancer Treat. Rev. 2020, 86, 102017. [Google Scholar] [CrossRef]
  22. Assoun, S.; Brosseau, S.; Steinmetz, C.; Gounant, V.; Zalcman, G. Bevacizumab in advanced lung cancer: State of the art. Future Oncol. 2017, 13, 2515–2535. [Google Scholar] [CrossRef]
  23. de Aguiar, R.B.; de Moraes, J.Z. Exploring the immunological mechanisms underlying the anti-vascular endothelial growth factor activity in tumors. Front. Immunol. 2019, 10, 1023. [Google Scholar] [CrossRef] [PubMed]
  24. Kim, H.R.; Sugawara, S.; Lee, J.; Kang, J.; Inui, N.; Hida, T.; Lee, K.H.; Yoshida, T.; Tanaka, H.; Yang, C.; et al. First-line nivolumab, paclitaxel, carboplatin, and bevacizumab for advanced non-squamous non-small cell lung cancer: Updated survival analysis of the ONO-4538-52/TASUKI-52 randomized controlled trial. Cancer Med. 2023, 12, 17061–17067. [Google Scholar] [CrossRef] [PubMed]
  25. Sugawara, S.; Lee, J.-S.; Kang, J.-H.; Kim, H.; Inui, N.; Hida, T.; Lee, K.; Yoshida, T.; Tanaka, H.; Yang, C.-T.; et al. Nivolumab with carboplatin, paclitaxel, and bevacizumab for first-line treatment of advanced nonsquamous non-small-cell lung cancer. Ann. Oncol. 2021, 32, 1137–1147. [Google Scholar] [CrossRef] [PubMed]
  26. Kanda, S.; Ohe, Y.; Goto, Y.; Horinouchi, H.; Fujiwara, Y.; Nokihara, H.; Yamamoto, N.; Yamamoto, T.; Tamura, T. Five-year safety and efficacy data from a phase Ib study of nivolumab and chemotherapy in advanced non-small-cell lung cancer. Cancer Sci. 2020, 111, 1933–1942. [Google Scholar] [CrossRef]
  27. Wu, L.; Yu, Y.; Zhou, J.; Wang, X.; Li, J.; Wang, Y. Mechanism of acquired resistance to nivolumab in lung squamous cell carcinoma: Case report and review of the literature. Immunotherapy 2020, 12, 957–964. [Google Scholar] [CrossRef]
  28. FDA. Opdivo-Highlights of Prescribing Information. 2022. Available online: http://www.fda.gov/medwatch (accessed on 9 September 2024).
  29. Barrios, D.M.; Do, M.H.; Phillips, G.S.; Postow, M.A.; Akaike, T.; Nghiem, P.; Lacouture, M.E. CME Part I: Immune checkpoint inhibitors to treat cutaneous malignancies. J. Am. Acad. Dermatol. 2020, 83, 1239. [Google Scholar] [CrossRef]
  30. Lin, X.; Kang, K.; Chen, P.; Zeng, Z.; Li, G.; Xiong, W.; Yi, M.; Xiang, B. Regulatory mechanisms of PD-1/PD-L1 in cancers. Mol. Cancer 2024, 23, 108. [Google Scholar] [CrossRef] [PubMed]
  31. FDA. Avastin-Highlights of Prescribing Information. 2014. Available online: http://www.fda.gov/medwatch (accessed on 9 September 2024).
  32. Haibe, Y.; Kreidieh, M.; El Hajj, H.; Khalifeh, I.; Mukherji, D.; Temraz, S.; Shamseddine, A. Resistance Mechanisms to Anti-angiogenic Therapies in Cancer. Front. Oncol. 2020, 10, 221. [Google Scholar] [CrossRef]
  33. Itatani, Y.; Kawada, K.; Yamamoto, T.; Sakai, Y. Resistance to Anti-Angiogenic Therapy in Cancer—Alterations to Anti-VEGF Pathway. Int. J. Mol. Sci. 2018, 19, 1232. [Google Scholar] [CrossRef]
  34. Hellmann, M.D.; Nathanson, T.; Rizvi, H.; Creelan, B.C.; Sanchez-Vega, F.; Ahuja, A.; Ni, A.; Novik, J.B.; Mangarin, L.M.; Abu-Akeel, M.; et al. Genomic Features of Response to Combination Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer. Cancer Cell 2018, 33, 843. [Google Scholar] [CrossRef]
  35. Nagasaki, J.; Ishino, T.; Togashi, Y. Mechanisms of resistance to immune checkpoint inhibitors. Cancer Sci. 2022, 113, 3303. [Google Scholar] [CrossRef] [PubMed]
  36. Vokes, N.I.; Pan, K.; Le, X. Efficacy of immunotherapy in oncogene-driven non-small-cell lung cancer. Ther. Adv. Med. Oncol. 2023, 15, 17588359231161408. [Google Scholar] [CrossRef]
  37. Zhao, Y.; Guo, S.; Deng, J.; Shen, J.; Du, F.; Wu, X.; Chen, Y.; Li, M.; Chen, M.; Li, X.; et al. VEGF/VEGFR-Targeted Therapy and Immunotherapy in Non-small Cell Lung Cancer: Targeting the Tumor Microenvironment. Int. J. Biol. Sci. 2022, 18, 3845–3858. [Google Scholar] [CrossRef]
  38. Melosky, B.; Juergens, R.; Hirsh, V.; McLeod, D.; Leighl, N.; Tsao, M.-S.; Card, P.B.; Chu, Q. Amplifying Outcomes: Checkpoint Inhibitor Combinations in First-Line Non-Small Cell Lung Cancer. Oncologist 2020, 25, 64–77. [Google Scholar] [CrossRef]
  39. Kato, Y.; Tabata, K.; Kimura, T.; Yachie-Kinoshita, A.; Ozawa, Y.; Yamada, K.; Ito, J.; Tachino, S.; Hori, Y.; Matsuki, M.; et al. Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS ONE 2019, 14, e0212513. [Google Scholar] [CrossRef]
  40. Herbst, R.S.; Arkenau, H.T.; Bendell, J.; Arrowsmith, E.; Wermke, M.; Soriano, A.; Penel, N.; Santana-Davila, R.; Bischoff, H.; Chau, I.; et al. Phase 1 Expansion Cohort of Ramucirumab Plus Pembrolizumab in Advanced Treatment-Naive NSCLC. J. Thorac. Oncol. 2021, 16, 289–298. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of action of nivolumab and bevacizumab.
Figure 1. Mechanisms of action of nivolumab and bevacizumab.
Medsci 13 00143 g001
Table 1. Evidence of nivolumab and bevacizumab studies with or without other therapeutic agents for NSCLC is summarized.
Table 1. Evidence of nivolumab and bevacizumab studies with or without other therapeutic agents for NSCLC is summarized.
Patient
Population
Study
Design
InterventionControlPrimary
Endpoint
Key ResultsRefs.
Treatment-
naïve stage HIB/IV
NSCLC
Randomized
1:1 trial
Nivolumab
with
bevacizumab
+ platinum-
based chemo
Placebo
+ chemo
OSOS: 30.8 vs.
24.7 months
(HR 0.74)
CI: 0.58–0.94
[24]
Treatment-
naïve stage IIIB/IV
NSCLC
Randomized,
double-
blind, 1:1 trial
Nivolumab
with
bevacizumab,
paclitaxel
+ platinum-based chemo
Placebo
+ chemo
PFSPFS: 12.1 vs.
8.1 months
(HR 0.56)
CI: 0.43–0.71
[25]
Japanese
patients
with
NSCLC
Phase IbNivolumab
with
bevacizumab,
paclitaxel
+ platinum-based chemo
NonePFS and OSPFS:
40.7 months,
OS:
28.5 months
CI: not reported
[26]
Advanced NSCLCMeta-
analysis of
phase III
clinical trials
Nivolumab + ipilimumab
with
bevacizumab
Chemo
alone
OSImproved OS in EGFR/
ALK mutated patients
CI: not reported
[38]
Table 2. Evidence from other ICI and anti-angiogenic-based approaches for NSCLC is summarized.
Table 2. Evidence from other ICI and anti-angiogenic-based approaches for NSCLC is summarized.
Patient
Population
Study DesignInterventionControlPrimary
Endpoint
Key ResultsRefs.
Metastatic non-squamous
NSCLC
Phase III clinical trialBevacizumab +
Carboplatin +
Paclitaxel
Atezolizumab + Bevacizumab
+ Carboplatin
+ Paclitaxel
PFS and OSMedian PFS: 8.3 months
(Intervention) vs. 6.8 months (Control) (HR:
0.59, p < 0.0001);
Median OS: 19.2 months
(Intervention) vs. 14.7 months (Control) (HR:
0.78, p = 0.02)
[18]
Metastatic
NSCLC
Preclinical and clinical studiesPembrolizumab
+ Lenvatinib
Not specifiedModulation of TMELenvatinib may decrease
TAMs and Tregs,
improving ICI efficacy (p < 0.01)
[39]
Metastatic
NSCLC
Open-label phase la/b trialPembrolizumab
+ Ramucirumab
Not specifiedPFS and OSMedian PFS: 9.3 months;
12-month and 18-month
PFS: 45%;
12-month and 18-month
OS: 73% and 64%
Respectively
(p value not reported)
[40]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Barney, T.; Thyagarajan, A.; Sahu, R.P. Combining Immune Checkpoint Inhibitors and Anti-Angiogenesis Approaches: Treatment of Advanced Non-Small Cell Lung Cancer. Med. Sci. 2025, 13, 143. https://doi.org/10.3390/medsci13030143

AMA Style

Barney T, Thyagarajan A, Sahu RP. Combining Immune Checkpoint Inhibitors and Anti-Angiogenesis Approaches: Treatment of Advanced Non-Small Cell Lung Cancer. Medical Sciences. 2025; 13(3):143. https://doi.org/10.3390/medsci13030143

Chicago/Turabian Style

Barney, Tate, Anita Thyagarajan, and Ravi P. Sahu. 2025. "Combining Immune Checkpoint Inhibitors and Anti-Angiogenesis Approaches: Treatment of Advanced Non-Small Cell Lung Cancer" Medical Sciences 13, no. 3: 143. https://doi.org/10.3390/medsci13030143

APA Style

Barney, T., Thyagarajan, A., & Sahu, R. P. (2025). Combining Immune Checkpoint Inhibitors and Anti-Angiogenesis Approaches: Treatment of Advanced Non-Small Cell Lung Cancer. Medical Sciences, 13(3), 143. https://doi.org/10.3390/medsci13030143

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop