9,871
Views
32
CrossRef citations to date
0
Altmetric
Drug Evaluation

Tislelizumab: an investigational anti-PD-1 antibody for the treatment of advanced non-small cell lung cancer (NSCLC)

&
Pages 1355-1364 | Received 03 Aug 2020, Accepted 05 Oct 2020, Published online: 21 Oct 2020

ABSTRACT

Introduction

Non-small cell lung cancer (NSCLC) accounts for most lung cancers worldwide and has a poor prognosis at later stages; programmed cell death protein-1 (PD-1) and programmed death-ligand 1 (PD-L1) inhibitors have provided promising new treatment approaches for these patients. Tislelizumab, an anti-PD-1 monoclonal antibody, was engineered to minimize binding to FcγR on macrophages to abrogate antibody-dependent phagocytosis, a mechanism of T-cell clearance and potential resistance to anti-PD-1 therapy. Tislelizumab has demonstrated clinical activity and is approved in China for treatment of previously treated classical Hodgkin lymphoma and previously treated metastatic PD-L1–high urothelial carcinoma.

Areas covered

This review summarizes the clinical efficacy, safety, and tolerability of tislelizumab in patients with NSCLC and examines the mechanism of action, pharmacokinetic, and pharmacodynamic profiles of tislelizumab.

Expert opinion

Tislelizumab has higher affinity to PD-1 than pembrolizumab and nivolumab, potentially due to its differential PD‑1 binding orientation. Tislelizumab demonstrated encouraging efficacy results, long duration of response, and a manageable safety profile across multiple clinical trials in advanced NSCLC. Ongoing trials of drug combinations (e.g. tislelizumab plus angiogenesis inhibitors, immune checkpoint inhibitors, or immune agonists) and examining efficacy across the severity of disease will provide opportunities to understand and feature tislelizumab in clinical practice.

1. Introduction

With approximately 2.1 million new cases and 1.8 million deaths each year, lung cancer is the leading cause of cancer-related death worldwide [Citation1]. Non-small cell lung cancer (NSCLC) accounts for the majority of lung cancers and has a poor prognosis at later stages [Citation2,Citation3]. The immune system plays an integral role in tumorigenesis and escape from immune surveillance is a recognized feature of cancer[Citation4]. Many new therapeutic strategies have focused on agents that block inhibitory T-cell immune checkpoints, including antibodies toward cytotoxic T-lymphocyte-associated protein 4 (CTLA4), programmed cell death protein-1 (PD-1), and programmed death-ligand 1 (PD-L1) [Citation5,Citation6,Citation7]. Despite limitations to access for some patients, PD-1/PD-L1 inhibitors continue to evolve to address the high mortality rate and are becoming part of the standard-of-care for many patients with NSCLC [Citation8].

2. Overview of the market

2.1. Checkpoint inhibitor monotherapy for previously treated NSCLC

Findings from key trials of patients with previously treated NSCLC receiving checkpoint inhibitor monotherapy are described below and included in . The global phase 2/3 KEYNOTE-010 study investigated different doses of pembrolizumab for patients with pretreated PD-L1–positive advanced NSCLC and showed improvements in median overall survival at both doses tested (pembrolizumab 2 mg/kg or 10 mg/kg) versus patients receiving docetaxel[Citation9]. Similarly, two global phase 2 trials (BIRCH and FIR) demonstrated that atezolizumab monotherapy had antitumor activity regardless of the line of therapy [Citation10,Citation11]. The potential benefits of nivolumab monotherapy for patients with pretreated advanced nonsquamous and squamous NSCLC was investigated in two large phase 3 trials (CheckMate 017 and CheckMate 057); both trials showed improved overall survival for patients receiving nivolumab versus those receiving docetaxel [Citation12,Citation13]. Additionally, durvalumab treatment resulted in longer overall survival than placebo in patients with stage III, unresectable NSCLC who did not have disease progression after concurrent radiotherapy in the PACIFIC trial [Citation14].

Table 1. Key immune checkpoint inhibitor monotherapy trials in patients with previously treated advanced NSCLC

2.2. Checkpoint inhibitors alone or in combination as first-line treatment of advanced NSCLC

Early indications that anti-PD-(L)1 therapy could provide clinical benefits as first-line treatment for patients with advanced NSCLC came from the phase 1 CheckMate 012 trial, in which 52 patients treated with first-line nivolumab monotherapy for advanced NSCLC had a median overall survival of 19.4 months after a median follow-up of 14.3 months [Citation15]. Because platinum-doublet chemotherapy is the historical first-line therapy for patients with advanced NSCLC without a targetable driver oncogene (e.g. EGFR, ALK, ROS1) [Citation16], many clinical studies combined PD-(L)1 inhibitors with chemotherapy as first-line treatment for advanced lung cancer and demonstrated improved survival [Citation17,Citation18,Citation19]; key clinical trials are included in . In KEYNOTE-024, pembrolizumab monotherapy showed longer overall survival in patients with tumor proportion score (TPS) ≥50%. In KEYNOTE-042, patients with TPS ≥1% treated with pembrolizumab monotherapy also demonstrated an improved overall survival at the end of the study across all PD-L1 expression subgroups [Citation20,Citation21]. Despite the statistically positive results at the end of KEYNOTE-042, chemotherapy initially out performed single-agent pembrolizumab across all TPS PD-L1 expression subgroups (≥50%, ≥20%; ≥1%). The overall positive result may have been driven by the survival of patients in the TPS PD-L1 ≥50% subgroup, which comprised nearly half of the study population. As such, it has been proposed that pembrolizumab monotherapy may not represent the best treatment strategy for patients with TPS PD-L1 scores of 1% to 49% [Citation22]. Furthermore, CheckMate 026 did not demonstrate a significant overall survival benefit for patients receiving first-line nivolumab versus those receiving chemotherapy (HR: 1.02; 95% CI: 0.80, 1.30) [Citation23].

Table 2. Key trials of first-line immune checkpoint inhibitors, as monotherapy and with chemotherapy, in patients with advanced NSCLC

2.3. Checkpoint inhibitors for first-line treatment of advanced squamous and nonsquamous NSCLC

Treatment regimens utilizing checkpoint inhibitors have also been investigated as therapy for nonsquamous and squamous NSCLC (). KEYNOTE-189 found that first-line pembrolizumab plus pemetrexed-platinum resulted in longer median overall survival versus the placebo plus chemotherapy combination group in patients with nonsquamous NSCLC[Citation27]. Atezolizumab plus chemotherapy also demonstrated longer median progression-free and overall survival compared with chemotherapy alone in patients with nonsquamous NSCLC in the IMpower150 and IMpower130 studies [Citation28,Citation29]. While patients with squamous NSCLC receiving pembrolizumab plus chemotherapy in KEYNOTE-407 also saw survival benefits compared with the placebo-chemotherapy group[Citation18], the results in patients with squamous NSCLC receiving atezolizumab plus chemotherapy were less straightforward. In IMpower131, overall survival benefits were not statistically significant with atezolizumab plus chemotherapy compared with chemotherapy alone, despite improved progression-free survival[Citation30].

2.4. Potential role of PD-L1 expression in the treatment of NSCLC

Several clinical trials have turned toward selecting patient populations using PD-L1 expression; however, to date, there is no universally accepted definition or standard assay for PD-L1–positivity. Tumor proportion score, PD-L1 tumor cell expression, and composite positive score, which includes tumor-infiltrating immune cells, have all been used to describe PD-L1 expression. This lack of a consistent definition has resulted in difficulties determining if PD-L1 is a universal predictive biomarker of response. Furthermore, the predictive value of PD-L1 expression is not always consistent within a single therapeutic agent. CheckMate 012 did not require PD-L1 expression for enrollment and indicated that first-line nivolumab monotherapy elicited durable responses in patients with advanced NSCLC[Citation15], but CheckMate 026, which only enrolled NSCLC patients with tumor cell PD-L1 expression ≥5%, did not demonstrate a significant survival benefit in patients receiving first-line nivolumab versus chemotherapy[Citation23]. Patients with PD-L1–positive NSCLC enrolled in KEYNOTE-024 and −042 studies were more consistent, as longer median overall survival was observed in both trials for patients receiving pembrolizumab monotherapy versus those receiving chemotherapy, including all PD-L1 expression levels examined in KEYNOTE-042 (TPS ≥50%, ≥20%, and ≥1%) [Citation20,Citation21].

PD-L1 expression may be more predictive of response to first-line checkpoint inhibitors in squamous versus nonsquamous NSCLC. In patients with squamous NSCLC receiving atezolizumab plus carboplatin and nab-paclitaxel during the IMpower131 trial, median progression-free and overall survival was longer in patients with high PD-L1 expression (PD-L1 expression on ≥50% of tumor cells or ≥10% of immune cells) versus those with lower levels of PD-L1 expression[Citation30]. However, patients with nonsquamous NSCLC receiving atezolizumab plus chemotherapy as part of the IMpower130 trial demonstrated clinical benefit versus chemotherapy regardless of PD-L1 expression[Citation29].

3. Introduction to tislelizumab

Tislelizumab (BGB-A317) is an anti-PD-1 monoclonal antibody that was engineered with a nullified Fc portion of the antibody to minimize binding to FcγR on macrophages in order to abrogate antibody-dependent phagocytosis, a mechanism of T-cell clearance and potential resistance to anti-PD-1 therapy [Citation31,Citation32]. Specifically, anti-PD-1 antibodies that bind FcγRs may mediate crosslinking between PD-1–positive T cells and FcγR-positive macrophages, which could induce macrophages to phagocytose active PD-1–positive T cells and possibly diminish cytolytic destruction of tumor cells [Citation31,Citation32]. As an antagonist to PD-L1/PD-L2–mediated cell signaling, tislelizumab leads to increased cytokine production and restoration of T-cell activation, resulting in immune-mediated tumor cell death[Citation31].

Box 1. Drug Summary

4. Chemistry

Tislelizumab is a humanized immunoglobulin G4 (IgG4) variant monoclonal antibody against the immune checkpoint-inhibitory receptor, PD-1 [Citation31].

5. Pharmacodynamics

Tislelizumab has a different binding orientation to PD‑1 versus pembrolizumab and nivolumab; the binding surface on PD‑1 for tislelizumab partially overlaps with that for pembrolizumab but differs significantly from that for nivolumab [Citation33]. With a disassociation constant (KD) of 0.15 nmol/L, tislelizumab binds to Gln75, Thr76, Asp77, and Arg86 of human PD-1 with high specificity and affinity [Citation33,Citation34]. Nivolumab and pembrolizumab do not require these epitopes to bind and have faster dissociation rates than tislelizumab. Tislelizumab has shown an approximately 50-fold and 100-fold slower dissociation rate compared with nivolumab and pembrolizumab, respectively [Citation33]. Studies with pembrolizumab and nivolumab have demonstrated that maximal receptor occupancy on circulating CD3 + T cells occurs at concentrations that are much lower than the clinical dose for nivolumab and pembrolizumab [Citation35,Citation36]. When the target-binding activity of tislelizumab was examined by fluorescence-activated cell sorting using a PD-1–expressing HuT78 cell line, the effective concentration for 50% of maximal activity was estimated to be 86 ng/mL.

6. Pharmacokinetics and metabolism

Tislelizumab is available as a sterile, injectable concentrate solution in a 100 mg per vial dosage form [Citation37]. After a single intravenous dose, tislelizumab demonstrated linear pharmacokinetics over a dose range of 0.5–10 mg/kg, including the recommended clinical dose of 200 mg every 3 weeks (Q3W) [Citation38]. After multiple-dose administration, steady-state concentration was achieved after 12 weeks with 200 mg Q3W. As the steady-state trough levels were greater than 30 μg/mL, tislelizumab is expected to reach maximal binding to PD-1.

The population pharmacokinetics of tislelizumab (0.5, 2.0, 5.0, or 10 mg/kg Q2W; 2.0 or 5.0 mg/kg Q3W; or 200 mg Q3W) were analyzed from 798 patients with advanced solid tumors or hematologic malignancies from two phase 1 and one phase 2 studies [Citation39]. Time-varying clearance was not observed; clearance and volume of distribution of the central compartment were estimated as 0.164 L/day and 2.92 L, respectively [Citation39]. The estimated terminal half-life of tislelizumab 200 mg Q3W was 16.8 ± 5.5 days [Citation40]. Covariate analysis of population pharmacokinetics indicated that dose adjustments were not needed based on age, sex, race, body weight, tumor size, serum albumin level, or immunogenicity status [Citation39]. The effect of renal or hepatic impairment on tislelizumab pharmacokinetics has not been directly evaluated, but population pharmacokinetics suggest that neither mild/moderate renal impairment nor mild hepatic impairment would require dose adjustments [Citation37].

No drug–drug interaction (DDI) studies have been conducted for tislelizumab thus far. Since no DDIs have been reported for other anti-PD-1/PD-L1 antibodies, the DDI potential of tislelizumab is expected to be low. Additionally, as the primary elimination pathways are protein catabolism via the reticuloendothelial system or target-mediated disposition, tislelizumab is not expected to induce or inhibit the major drug-metabolizing CYP pathways.

7. Clinical efficacy

Initial antitumor activity of single-agent tislelizumab was assessed in previously treated patients with advanced solid tumors in two early phase clinical studies (). In a global, phase 1A/1B first-in-human study (BGB-A317-001; NCT02407990), tislelizumab treatment in all patients (n = 451) resulted in a median overall survival of 10.3 months (95% CI: 8.5, 11.6) with a median duration of study follow-up of 8.6 months (range: 0.1, 40.8) [Citation40]. Of the 49 evaluable patients with NSCLC treated with tislelizumab 200 mg Q3W in the phase 1B dose-expansion phase, an objective response rate of 12.2% (95% CI: 4.6, 24.8) was observed, despite a high proportion of patients having received prior systemic drug therapy (n = 44, 90%) [Citation40,Citation41]. Clinical benefit was observed in both Asian and non-Asian patients [Citation40], as well in patients with PD-L1–positive (≥25% of tumor cells exhibiting PD-L1 membrane) staining and PD-L1–negative (<25% PD-L1 tumor cell expression) tumors [Citation41]. Across all patients from an open-label phase 1/2 study conducted in China (BGB-A317-102; NCT04068519), treatment with tislelizumab resulted in a median overall survival of 11.5 months (95% CI: 9.1, 15.0) with a median follow-up of 12.2 months [Citation42]. Among previously treated patients with NSCLC (n = 56), the objective response rate was 18% (95% CI: 8.9, 30.4), with 31 patients achieving confirmed partial response (n = 10; 18%) or stable disease (n = 21; 38%) [Citation42]. Median progression-free survival for patients with NSCLC was 4.0 months (95% CI: 2.1, 8.1) and at the time of data cutoff, median overall survival had not been reached [Citation42]. Consistent with the first-in-human study, responses were observed in patients with NSCLC regardless of PD-L1 status[Citation42].

Table 3. Key tislelizumab trials enrolling patients with lung cancera

Tislelizumab plus chemotherapy as first-line treatment for NSCLC has been evaluated in three clinical studies. In an open-label phase 2 trial in China (BGB-A317-206; NCT03432598), eligible patients with advanced/metastatic nonsquamous NSCLC, squamous NSCLC, or extensive-stage small cell lung cancer (SCLC) received tislelizumab plus 4–6 cycles of platinum-doublet. The nonsquamous cohort received tislelizumab with pemetrexed plus platinum Q3W for four cycles followed by pemetrexed maintenance. Patients with squamous NSCLC received tislelizumab plus either paclitaxel plus platinum (Cohort A) or gemcitabine plus platinum (Cohort B) Q3W, and patients in the SCLC cohort received tislelizumab plus etoposide plus platinum Q3W[Citation43]. Median progression-free survival estimates were 9.0 months (nonsquamous), 7.0 months (squamous Cohort A), and 6.9 months (SCLC); median progression-free survival was not reached for squamous NSCLC Cohort B. Despite the long survival follow-up times (17.4 months, nonsquamous; 18.3 months, squamous Cohort A; 18.1 months, squamous Cohort B; 15.3 months, SCLC), median overall survival was not reached in all cohorts, except for SCLC (15.6 months). The 1-year overall survival rates were 88% (nonsquamous), 93% (squamous Cohort A), 80% (squamous Cohort B), and 76% (SCLC).

In an open-label, randomized phase 3 trial in China (RATIONALE 307; BGB-A317-307; NCT03594747) examining first-line treatment for advanced squamous NSCLC (n = 360), the addition of tislelizumab to paclitaxel plus carboplatin or nab-paclitaxel plus carboplatin significantly improved progression-free survival when compared with paclitaxel plus carboplatin only (HR: 0.52 and 0.48, respectively) [Citation44]. Median progression-free survival was 7.6 months for both tislelizumab-containing arms; median progression-free survival for chemotherapy alone was 5.5 months. The objective response rate was 72.5% for tislelizumab with paclitaxel plus carboplatin and 74.8% for patients treated with tislelizumab combined with nab-paclitaxel plus carboplatin, respectively; the objective response rate for chemotherapy alone was 49.6%. Median duration of response was 8.2 months for patients receiving tislelizumab combined with paclitaxel plus carboplatin, 8.6 months for tislelizumab combined with nab-paclitaxel plus carboplatin, and 4.2 months for chemotherapy alone. There was no association between PD-L1 expression levels (<1%, 1–49%, and ≥50%) and progression-free survival or clinical response. Together, these data represent the second clinical trial to demonstrate positive clinical outcomes in patients with squamous NSCLC.

In April 2020, it was announced that an interim analysis of an open-label phase 3 trial (RATIONALE 304; BGB-A317-304; NCT03663205) evaluating first-line tislelizumab plus pemetrexed and platinum-based chemotherapy for patients with nonsquamous NSCLC met its primary endpoint, demonstrating a statistically significant improvement in progression-free survival, as assessed by an independent review committee, versus chemotherapy alone [Citation45]. The safety profile of tislelizumab plus pemetrexed and platinum-based chemotherapy was consistent with the known risks of each study treatment component; no new safety signals were identified.

8. Safety and tolerability

As monotherapy, the adverse events reported in patients treated with tislelizumab were manageable and generally mild-to-moderate in severity [Citation40,Citation42]. In the first-in-human study, median treatment duration of tislelizumab was 2.8 months (range: 0.13–43.7) and 65 patients (14.4%) received treatment for ≥12 months [Citation40]. Treatment-related adverse events led to discontinuation in 5.3% of patients [Citation40]. In the open-label phase 1/2 study, median treatment duration of patients receiving tislelizumab 200 mg was 18 weeks (range: 0.9, 95.7) [Citation42]. Treatment-emergent adverse events led to discontinuation in 8% of patients and the only treatment-related adverse events that led to discontinuation in more than one patient were autoimmune myocarditis, lung infection, central nervous system metastases, and death (n = 2 each) [Citation42]. In this study, most adverse events considered related to tislelizumab by the investigator were generally of grade ≤2 severity and no new immune-related safety signals were observed [Citation42].

In the phase 2 study of first-line tislelizumab plus chemotherapy in patients with lung cancer (n = 54), six patients (11%) discontinued tislelizumab due to adverse events (pneumonitis [n = 2], dyspnea, increased conjugated bilirubin, decreased platelet count, myocarditis, immune-mediated hepatitis, post-procedural discomfort, and rhabdomyolysis [n = 1 each]) [Citation43].

In the phase 3 study of first-line tislelizumab plus chemotherapy for squamous NSCLC, the most commonly reported grade ≥3 adverse events were hematologic toxicities (e.g. neutropenia), which is consistent with adverse events known to be associated with chemotherapy. Adverse events leading to discontinuation of any treatment were reported in 12.5% of patients receiving tislelizumab plus paclitaxel and carboplatin, and in 15.4% of patients receiving paclitaxel and carboplatin [Citation44]. Tislelizumab had no impact on the tolerability of chemotherapy and did not affect exposure duration, numbers and cycles of administration, or dose intensity of chemotherapy.

9. Regulatory affairs

In December 2019, tislelizumab received conditional approval from China’s National Medical Products Administration (NMPA) for patients with relapsed/refractory classical Hodgkin lymphoma who have received ≥2 prior therapies. In April 2020, the NMPA also approved tislelizumab for patients with previously treated locally advanced or metastatic urothelial carcinoma with PD-L1–high expression [Citation37]. The NMPA accepted for review two supplemental new drug applications for tislelizumab plus chemotherapy as first-line treatment for advanced squamous (April 2020) and nonsquamous (June 2020) NSCLC. In addition to the three previously described studies, tislelizumab efficacy and safety are being assessed as second- and third-line treatments for patients with advanced NSCLC (RATIONALE 303; BGB-A317-303; NCT03358875).

10. Conclusions

Tislelizumab was generally well tolerated with no unexpected safety concerns observed in patients with advanced NSCLC. Promising efficacy was observed regardless of ethnicity and PD-L1 expression in patients receiving tislelizumab as monotherapy in heavily pretreated NSCLC patients and with chemotherapy as first-line treatment for squamous and nonsquamous NSCLC. The results support further investigation of tislelizumab in NSCLC.

11. Expert opinion

Anti-PD-1/PD-L1 antibodies, either alone or with chemotherapy, have significantly improved clinical benefit in patients with squamous and nonsquamous advanced NSCLC, both as first-line treatment and as a later line of therapy. However, a significant proportion of patients remain unresponsive or subsequently progress following initial treatment. Therefore, novel checkpoint inhibitor therapies and new approaches, such as bispecific antibodies (e.g. M7824), which have the potential to simultaneously inhibit the PD-(L)1 pathway with inhibition of other cellular processes, have also been investigated for the treatment of NSCLC. To develop effective treatment regimens, more knowledge of NSCLC disease biology is needed, including a better understanding of de novo/emerging mechanisms of resistance, the biomarkers and phenotypes associated with each, and novel treatment approaches to overcome any resistance that develops.

In the current age of personalized medicine, identification of predictive biomarkers of response may enable better-informed treatment decisions and provide consistency in immune checkpoint inhibitor selection. As clinicians strive to achieve better patient outcomes, additional predictive indicators of response to immunotherapy outside of PD-L1 expression, such as tumor mutational burden (TMB), blood TMB (bTMB), and circulating tumor DNA (ctDNA), are being investigated. High TMB is indicative of genomic instability and has been correlated with responses to immunotherapy and improved efficiency of immune checkpoint inhibitor treatment [Citation46–48]. However, the predictive value on long-term survival in patients with NSCLC requires further evaluation. Because many patients with advanced cancer cannot provide enough tumor tissue for molecular testing[Citation49], noninvasive approaches utilizing advances in DNA sequencing technology are attractive options. As such, bTMB, which represents a rapid blood-based assay that may be less impacted by intratumor heterogeneity than tissue-based TMB, has garnered much interest [Citation49]. Additionally, examination of key tumor-related genes via ctDNA has introduced the possibility of achieving earlier cancer detection and improved therapeutic outcomes by enabling early intervention [Citation50].

RATIONALE 307 was only the second trial of a checkpoint inhibitor to meet its full primary endpoint in patients with advanced/metastatic squamous NSCLC; in KEYNOTE-407, patients with metastatic squamous NSCLC receiving pembrolizumab platinum-containing chemotherapy reported significantly prolonged overall survival and progression-free survival (dual primary endpoints) compared with chemotherapy alone [Citation18]. Tislelizumab is being investigated as a single-agent in second- and third-line settings and with chemotherapy as first-line treatment for advanced NSCLC. Additionally, tislelizumab plus chemotherapy is being examined as neoadjuvant treatment and single-agent tislelizumab is being explored as adjuvant maintenance therapy. Tislelizumab was engineered to minimize binding to FcγR on macrophages and may result in prolonged antitumor effects versus therapies where FcγR is engaged. Tislelizumab has demonstrated encouraging efficacy results, long duration of response, a manageable safety profile across multiple clinical trials in advanced NSCLC, and responses were comparable across both Asian and non-Asian patients [Citation40], as well as being comparable regardless of PD-L1 expression [Citation40,Citation42]. The favorable tolerability profile and durable efficacy of tislelizumab in patients with NSCLC suggests that tislelizumab may be appropriate as a checkpoint inhibitor backbone for combination therapies with chemotherapy or other novel agents. Longer survival follow-up from the current clinical trials should allow for an opportunity to examine the impact of tislelizumab on overall survival.

Within the next 5 years, tislelizumab has the potential to bring clinical benefit to patients with NSCLC in all phases of treatment. The safety profile of tislelizumab suggests that it may be an ideal candidate as the backbone of combination therapy with chemotherapy or targeted treatments, which could help to overcome acquired resistance to immunotherapy. Combining tislelizumab with other anticancer agents is currently being investigated in multiple treatment settings and may further improve the survival benefits already observed with tislelizumab.

Multiple PD-(L)1 therapies are available for the treatment of squamous and nonsquamous NSCLC, so it is important to take a holistic approach and to evaluate each agent based on efficacy, safety and tolerability, ease of administration, quality of life and patient-reported outcomes, and cost-effectiveness. Due to the high prevalence of NSCLC, significant economic burden of this illness, and the associated impact on societal and healthcare costs, real-world evidence, and comparative cost-effectiveness analyses will be needed. The value of immune checkpoint inhibitors in novel combinations for areas of unmet medical need (e.g. patients with brain metastases, those who have progressed after prior EGFR tyrosine kinase inhibitor or an immune checkpoint inhibitor) is also an area of ongoing investigation. With a manageable safety profile and long-lasting responses regardless of PD-L1 expression levels, tislelizumab looks to build upon the transformative clinical outcomes brought about by checkpoint inhibitor therapy.

Article highlight

  • Tislelizumab is Tislelizumab is a humanized, IgG4 monoclonal antibody with high affinity and binding specificity for programmed cell death protein-1 (PD-1) that was engineered to minimize binding to FcγR on macrophages in order to abrogate antibody-dependent phagocytosis, a mechanism of T-cell clearance and potential resistance to anti-PD-1 therapy

  • When administered as either first-line or as a later line of therapy, anti-PD-1/PD-L1 antibodies havesignificantly improved clinical outcomes in patients with squamous and nonsquamous advanced NSCLC alone and when combined with chemotherapy

  • As a single-agent, tislelizumab has demonstrated encouraging efficacy regardless of PD-L1 expression levels with a manageable safety/tolerability profile across multiple clinical trials in advanced NSCLC

  • In combination with chemotherapy, tislelizumab has shown prolonged survival and durable clinical responses; the safety/tolerability profile was consistent with the known risks of each study treatment component with no new safety signals

  • Ongoing trials examining efficacy across the severity of disease will provide additional opportunities to understand and feature tislelizumab in clinical practice

  • With a manageable safety profile and long-lasting responses regardless of PD-L1 expression levels, tislelizumab looks to build upon the transformative clinical outcomes brought about by checkpoint inhibitor therapy

Declaration of interest

Yi-Long Wu has received speaker fees from AstraZeneca, Boehringer Ingelheim, BMS, Eli Lilly, MSD, Pfizer, Roche, and Sanofi. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer Disclosures

Peer reviewers on this manuscript have no relevant financial relationships or otherwise to disclose.

Acknowledgments

Financial support for the development of this manuscript was provided by BeiGene, Ltd. This included medical writing and editorial assistance under the authors’ guidance, and was provided by Stephan Lindsey PhD and Elizabeth Hermans PhD of OPEN Health Medical Communications (Chicago, IL, USA).

Additional information

Funding

This article is funded by BeiGene, Ltd.

References

  • Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018 Nov;68(6):394–424.
  • D’Addario G, Fruh M, Reck M, et al. Metastatic non-small-cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010 May;21(suppl 5):v116–119.
  • Blandin Knight S, Crosbie PA, Balata H, et al. Progress and prospects of early detection in lung cancer. Open Biol. 2017 Sep;7(9):170070.
  • Smida T, Bruno TC, Stabile LP. Influence of estrogen on the NSCLC microenvironment: a comprehensive picture and clinical implications. Front Oncol. 2020;10:137.
  • Akinleye A, Rasool Z. Immune checkpoint inhibitors of PD-L1 as cancer therapeutics. J Hematol Oncol. 2019 Sep 5;12(1):92.
  • McDermott DF, Atkins MB. PD-1 as a potential target in cancer therapy. Cancer Med. 2013 Oct;2(5):662–673.
  • Weber J. Overcoming immunologic tolerance to melanoma: targeting CTLA-4 with ipilimumab (MDX-010). Oncologist. 2008;13(suppl 4):16–25.
  • Thallinger C, Fureder T, Preusser M, et al. Review of cancer treatment with immune checkpoint inhibitors: current concepts, expectations, limitations and pitfalls. Wien Klin Wochenschr. 2018 Feb;130(3–4):85–91.
  • Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016 Apr 9;387(10027):1540–1550.
  • Peters S, Gettinger S, Johnson ML, et al. Phase II trial of atezolizumab as first-line or subsequent therapy for patients with programmed death-ligand 1-selected advanced non-small-cell lung cancer (BIRCH). J Clin Oncol. 2017 Aug 20;35(24):2781–2789.
  • Spigel DR, Chaft JE, Gettinger S, et al. FIR: efficacy, safety, and biomarker analysis of a phase II open-label study of atezolizumab in PD-L1-selected patients with NSCLC. J Thorac Oncol. 2018 Nov;13(11):1733–1742.
  • Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015 Jul 9;373(2):123–135.
  • Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015 Oct 22;373(17):1627–1639.
  • Antonia SJ, Villegas A, Daniel D, et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med. 2018 Dec 13;379(24):2342–2350.
  • Gettinger S, Rizvi NA, Chow LQ, et al. Nivolumab monotherapy for first-line treatment of advanced non-small-cell lung cancer. J Clin Oncol. 2016 Sep 1;34(25):2980–2987.
  • Shi Y, Sun Y, Yu J, et al. China experts consensus on the diagnosis and treatment of advanced stage primary lung cancer (2016 version). Asia Pac J Clin Oncol. 2017 Feb;13(1):87–103.
  • Horn L, Mansfield AS, Szczesna A, et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med. 2018 Dec 6;379(23):2220–2229.
  • Paz-Ares L, Luft A, Vicente D, et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med. 2018 Nov 22;379(21):2040–2051.
  • Gandhi L, Rodriguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018 May 31;378(22):2078–2092.
  • Reck M, Rodriguez-Abreu D, Robinson AG, et al. Updated analysis of KEYNOTE-024: pembrolizumab versus platinum-based chemotherapy for advanced non-small-cell lung cancer with PD-L1 tumor proportion score of 50% or greater. J Clin Oncol. 2019 Mar 1;37(7):537–546.
  • Mok TSK, Wu YL, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet. 2019 May 4;393(10183):1819–1830.
  • Mountzios G, Remon J, Novello S, et al. Position of an international panel of lung cancer experts on the decision for expansion of approval for pembrolizumab in advanced non-small-cell lung cancer with a PD-L1 expression level of >/=1% by the USA food and drug administration. Ann Oncol. 2019 Nov 1;30(11):1686–1688.
  • Carbone DP, Reck M, Paz-Ares L, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017 Jun 22;376(25):2415–2426.
  • Spigel D, de Marinis F, Giaccone G, et al. IMpower110: interim overall survival analysis of a phase III study of atezolizumab vs platinum-based chemotherapy as first-line treatment in PD-L1-selected NSCLC. Ann Oncol. 2019;30(suppl 5):v851–934.
  • Rizvi NA, Cho BC, Reinmuth N, et al. Durvalumab with or without tremelimumab vs standard chemotherapy in first-line treatment of metastatic non-small cell lung cancer: the MYSTIC phase 3 randomized clinical trial. JAMA Oncol. 2020 May 1;6(5):661–674.
  • Reck M, Ciuleanu TE, Cobo Dols M, et al. Nivolumab (NIVO) + ipilimumab (IPI) + 2 cycles of platinum-doublet chemotherapy (chemo) vs 4 cycles chemo as first-line (1L) treatment (tx) for stage IV/recurrent non-small cell lung cancer (NSCLC): checkMate 9LA. J Clin Oncol. 2020;38(suppl):Abstract 9501.
  • Gadgeel S, Rodriguez-Abreu D, Speranza G, et al. Updated analysis from KEYNOTE-189: pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous non-small-cell lung cancer. J Clin Oncol. 2020 May 10;38(14):1505–1517.
  • Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018 Jun 14;378(24):2288–2301.
  • West H, McCleod M, Hussein M, et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019 Jul;20(7):924–937.
  • Jotte R, Cappuzzo F, Vynnychenko I, et al. Atezolizumab in combination with carboplatin and nab-paclitaxel in advanced squamous NSCLC (IMpower131): results from a randomized phase III trial. J Thorac Oncol. 2020 Apr 14;S1556–0864(20):30292–30296.
  • Zhang T, Song X, Xu L, et al. The binding of an anti-PD-1 antibody to FcgammaRIota has a profound impact on its biological functions. Cancer Immunol Immunother. 2018 Jul;67(7):1079–1090.
  • Dahan R, Sega E, Engelhardt J, et al. FcγRs modulate the anti-tumor activity of antibodies targeting the PD-1/PD-L1 axis. Cancer Cell. 2015 Sep 14;28(3):285–295.
  • Feng Y, Hong Y, Sun H, et al. The molecular binding mechanism of tislelizumab, an investigational anti-PD-1 antibody, is differentiated from pembrolizumab and nivolumab. Proceedings of the 110th Annual Meeting of the American Association for Cancer Research. Atlanta, GA: American Association of Cancer Research; 2019. p. Abstract 4048.
  • Kaplon H, Reichert JM. Antibodies to watch in 2019. MAbs. 2019 Feb/Mar;11(2):219–238.
  • Patnaik A, Kang SP, Rasco D, et al. Phase I study of pembrolizumab (MK-3475; anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. Clin Cancer Res. 2015 Oct 1;21(19):4286–4293.
  • Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012 Jun 28;366(26):2443–2454.
  • Lee A, Keam SJ. Tislelizumab: first approval. Drugs. 2020 Mar 18;80(6):617–624.
  • Desai J, Markman B, Sandhu S, et al. Updated safety, efficacy, and pharmacokinetics (PK) results from the phase I study of BGB-A317, an anti- programmed death-1 (PD-1) mAb in patients with advanced solid tumors. J Immunother Cancer. 2016;4(Suppl1):AbstractP154.
  • Wu C-Y, Tang T, Liu L, et al. Population pharmacokinetics of tislelizumab in patients with advanced tumors. Ann Oncol. 2019;30(suppl 5):183.
  • Desai J, Deva S, Lee JS, et al. Phase IA/IB study of single-agent tislelizumab, an investigational anti-PD-1 antibody, in solid tumors. J Immunother Cancer. 2020 Jun;8(1):e000453.
  • Barlow P, Jameson M, Lin -C-C, et al. Single-agent tislelizumab, an anti-PD-1 antibody: results from a phase 1 expansion cohort in NSCLC patients.Poster presented at: the 20th World Congress on Lung Cancer; September 07–10, 2019; Barcelona, Spain.
  • Shen L, Guo J, Zhang Q, et al. Tislelizumab in Chinese patients with advanced solid tumors: an open-label, non-comparative, phase 1/2 study. J Immunother Cancer. 2020 Jun;8:1.
  • Wang Z, Zhao J, Ma Z, et al. A phase 2 study of tislelizumab in combination with platinum-based chemotherapy as first-line treatment for advanced lung cancer in chinese patients. Lung Cancer. 2020 Sep;147:259–268.
  • Wang J, Lu S, Hu C et al. Updated analysis of tislelizumab plus chemotherapy vs chemotherapy alone as first-line treatment of advanced squamous non-small cell lung cancer (SQ NSCLC). Presented at: 2020 European Society for Medical Oncology Meeting; 19–21 September, 2020; Virtual Congress.
  • BeiGene announces that the phase 3 trial of tislelizumab combined with chemotherapy in patients with first-line non-squamous non-small cell lung cancer met the primary endpoint of progression-free survival at interim analysis. [cited 2020 Jul 09]. Available from: http://ir.beigene.com/news-releases/news-release-details/beigene-announces-phase-3-trial-tislelizumab-combined
  • Hellmann MD, Ciuleanu TE, Pluzanski A, et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018 May 31;378(22):2093–2104.
  • Wang C, Liang H, Lin C, et al. Molecular subtyping and prognostic assessment based on tumor mutation burden in patients with lung adenocarcinomas. Int J Mol Sci. 2019 Aug 30;20(17):4251.
  • Wu Y, Xu J, Du C, et al. The predictive value of tumor mutation burden on efficacy of immune checkpoint inhibitors in cancers: a systematic review and meta-analysis. Front Oncol. 2019;9:1161.
  • Gandara DR, Paul SM, Kowanetz M, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat Med. 2018 Sep;24(9):1441–1448.
  • Abbosh C, Birkbak NJ, Swanton C. Early stage NSCLC - challenges to implementing ctDNA-based screening and MRD detection. Nat Rev Clin Oncol. 2018 Sep;15(9):577–586.