Next Article in Journal
In Utero Exposure to Hormonal Contraception and Mortality in Offspring with and without Cancer: A Nationwide Cohort Study
Next Article in Special Issue
Small Cell Lung Cancer: Emerging Targets and Strategies for Precision Therapy
Previous Article in Journal
The Emerging Role of Metformin in the Treatment of Hepatocellular Carcinoma: Is There Any Value in Repurposing Metformin for HCC Immunotherapy?
Previous Article in Special Issue
Emerging Targeted Therapies in Advanced Non-Small-Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Arising Novel Agents in Lung Cancer: Are Bispecifics and ADCs the New Paradigm?

Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA 910102, USA
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(12), 3162; https://doi.org/10.3390/cancers15123162
Submission received: 18 May 2023 / Revised: 5 June 2023 / Accepted: 7 June 2023 / Published: 13 June 2023

Abstract

:

Simple Summary

Lung cancer is the leading cause of cancer-related death worldwide according to the World Health Organization. Non-small cell lung cancer makes up the majority of cases. Immunotherapy with immune checkpoint inhibitors and targeted therapy with tyrosine kinase inhibitors and other molecular targeted agents significantly changed the treatment landscape and overall survival. Unfortunately, resistance to these treatments develops, and there is a need to identify additional innovative therapies that can overcome treatment resistance. Advancements in biomedical engineering and technology allowed for the development of novel agents, capable of delivering effective treatment directly to tumor cells. These agents include antibody drug conjugates and bispecific antibodies which have various targets and mechanisms of action, discussed in depth in this review.

Abstract

Lung cancer is one of the most common cancers with the highest mortality. Non-small cell lung cancer (NSCLC) contributes to around 85% of lung cancer diagnoses (vs. 15% for small cell lung cancer). The treatment of NSCLC has vastly changed in the last two decades since the development of immunotherapy and targeted therapy against driver mutations. As is the nature of malignancy, cancer cells have acquired resistance to these treatments prompting an investigation into novel treatments and new targets. Bispecific antibodies, capable of targeting multiple substrates at once, and antibody–drug conjugates that can preferentially deliver chemotherapy to tumor cells are examples of this innovation. From our initial evaluation, both treatment modalities appear promising.

1. Introduction

Lung cancer is the leading cause of cancer-related death worldwide according to the WHO [1]. Non-small cell lung cancer (NSCLC) contributes to around 85% of lung cancer diagnoses (vs. 15% for small cell lung cancer), which is further categorized into adenocarcinoma 78% and squamous 18% [2]. Since the early 2000s, the treatment options for NSCLC have exponentially increased after the discovery of immune checkpoint inhibitors and targetable ‘driver mutations’ [2,3]. Immune checkpoint inhibitors, pembrolizumab, atezolizumab, and cemiplimab were proven effective first-line agents in programmed cell death ligand 1 [PD-L1] positive metastatic NSCLC after the respective phase 3 trials [4,5,6]. Combination PD-L1 and cytotoxic T-lymphocyte-associated protein 4 [CTLA-4] inhibitors, nivolumab and ipilimumab were also proven in the first line compared to standard-of-care chemotherapy regardless of PD-L1 status [7].
The first successful targets of ‘driver mutations’ were epidermal growth factor receptor (EGFR)-activating mutations, most commonly deletions in exon 19 or point mutation L858R in exon 21, found in approximately 15% of adenocarcinomas [2,8,9,10]. Tumors with these mutations demonstrated significant sensitivity to tyrosine kinase inhibitors (TKIs; gefitinib and erlotinib) prompting investigation in several clinical trials [OPTIMAL, EURTAC, ENSURE, etc.]. These trials confirmed the superior ORR and median PFS of these first-generation TKIs versus standard-of-care chemotherapy and established their use as first-line therapy [11,12,13,14,15].
Genetic alterations developed, notably the T790M mutation, which conferred resistance to the EGFR first-generation agents [16]. From the landmark FLAURA trial, third-generation TKI osimertinib demonstrated effectiveness as first-line treatment with improved median PFS and OS compared to the first-generation agents, thereby becoming the new standard of care [17]. Additionally, numerous other driver mutations have entered the standard of care in first- and second-line treatment, notably ALK, ROS1, BRAF V600E, MET Ex14 skipping, RET, HER2, NTRK, and, most recently, KRAS G12C [18,19,20,21,22,23,24,25,26]. Unfortunately, resistance either through target-dependent (changes in the structure of the tyrosine kinase/target preventing inhibition) or target-independent (upregulation of bypass signaling pathways) emerged [27]. This resistance to treatment with molecular targeted inhibitors and immune checkpoint blockade prompted researchers to investigate mechanisms and pathways to circumvent it. This review will provide an analysis of some of these new treatments and pathway targets.

2. Bispecific Antibodies

The concept of antibodies with the capability of targeting and utilizing multiple substrates (Bispecific, Trispecific) which could then be further enhanced to utilize T cells, so-called Bispecific T cell Engagers (BiTE), is relatively new in the context of cancer treatment. It was not until 1984 when BiTEs were first shown to employ T cells to prevent the growth of tumor cells in vivo [28]. Success in clinical trials was limited following the initial discovery, but molecular modifications discovered from preclinical data eventually translated into clinical results [29]. The first successful use of bispecific antibodies that did not utilize T cells was seen in end-stage Hodgkin’s lymphoma where one patient treated with an NK cell activating CD16/CD30 bispecific antibody achieved a complete remission while another achieved a partial remission from the study group of 15 patients [30]. Since then, advances in protein engineering and molecular techniques prompted a variety of formulations and targets but the most common remains bispecific antibodies that redirect effector immune cells [T cells] to target malignant cells [BiTE] [31]. The quick adoption of this treatment modality was not universal across the spectrum of malignancies, but interest has peaked recently, and lung cancer is no exception.

2.1. C-Met Targeting

While the prevalence of EGFR resistance in NSCLC is increasing since the introduction and widespread use of TKIs, bispecific antibodies may provide a potential solution. EGFR mutations with insertions in exon 20, except A763_Y764insFQEA, have demonstrated very low response rates (<10%) to common EGFR TKIs (osimertinib, erlotinib, etc.), which is thought to be related to alterations that prevent TKI binding [32,33]. Additionally, MET gene amplification leads to the upregulation of cMet, and the subsequent pathway allows cancer cells to bypass EGFR signaling for cell survival, thereby creating another avenue of resistance to our current EGFR-targeted treatments [34]. The CHRYSALIS trial, a phase I trial dose-escalation/dose-expansion study, sought to focus on both resistance patterns with amivantamab, a dual EGFR and cMET bispecific antibody [35] (Figure 1). The initial data were promising with a PFS of 8.3 months [95% CI, 6.5 to 10.9] and an ORR of 40% [95% CI, 29 to 51] [34] (Table 1). The drug appears to have tolerable side effects which include infusion reactions most commonly seen during the first cycle, 49% grade 2/3 rash, and 20% grade 2/3 paronychia [35].
Additional investigation into amivantamab is currently underway including MARIPOSA1, a phase 3 trial evaluating the combination of amivantamab and lazertinib versus osimertinib in the first-line setting [36]. There are also early studies in the preclinical and phase 1 settings attempting to identify additional targets and combination agents involving cMet signaling. One such example is AZD-9592, a bispecific antibody that targets both EGFR and c-Met but is also conjugated to a topoisomerase inhibitor, which allows for both cytotoxic effects and receptor signaling blockade, currently recruiting in a phase 1 trial (NCT05647122) (Table 1).

2.2. HER2/HER3 Targeting

After human epidermal growth factor 2 (HER2) targeting was utilized in breast cancer successfully, similar attempts were made in lung cancer. Bispecific antibodies were once again implemented to target multiple facets of cell regulation. Zenocutuzumab, a bispecific antibody designed with one Fab arm that binds to HER2, allows the other Fab arm targeting human epidermal growth factor 3 (HER3) to be in a position to prevent NRG1 binding to HER3, thereby blocking activation and subsequent cell signaling [37] (Figure 1). This drug was first proven in pre-clinical models of lung, breast, pancreas, and ovarian cancers with neuregulin-1 (NRG1) fusions in both in vitro and in vivo settings [38]. In the same study, a patient who had progressed on multiple lines of therapy with CD74-NRG1 NSCLC responded quickly with a partial response to treatment [38]. Currently, there is a phase II trial evaluating this drug in both NSCLC and pancreatic cancer in patients with NRG1 fusions (NCT02912949) (Table 1). Additional targeted therapy directed towards HER2/HER3 has entered the forefront of NSCLC treatment and will be discussed in later sections.

2.3. PD-1/CTLA-4 Targeting

Immune checkpoint inhibitors have been at the center of NSCLC treatment for the last decade. Data from clinical trials in lung cancer, as well as melanoma and renal cell carcinoma, demonstrate that dual CTLA-4 and PD-L1 inhibition improves overall survival [7,39,40,41]. Unfortunately, there were increased rates of immune-related adverse events at the optimal effective treatment dose of CTLA-4/PD-L1 inhibitors [42]. This left a role for novel agents that are able to target both PD-L1 and CTLA-4 while maintaining tolerable toxicity. MEDI5752, a bispecific antibody of PD-1 and CTLA4, was specifically designed to bind CTLA-4- in PD-1-positive T cells allowing a higher level of T cell proliferation [43]. The Initial results of the phase 1b/II trial of MEDI5752 plus chemotherapy (carboplatin and pemetrexed) vs. pembrolizumab plus chemotherapy in the first line were presented at ESMO in September 2022; ORR was 50.0 [95% CI 27.2–72.8] with the study drug versus 47.6 [95% CI 25.7–70.2] in the pembrolizumab group [44]. Notable separation in ORR was seen in the PD-L1 < 1% group with ORR of 55.6 [95% CI 21.2–86.3] with the study drug and 30.0 [95% CI 6.7–65.2] (NCT03530397) (Table 1). Phase III trial data of this novel agent may provide additional insight into the efficacy and safety of this treatment modality. Another PD-L1 targeting agent, INBRX-105, a bispecific against PD-L1 and human 4-1BB receptor (CD137), is being investigated in a phase II study, used in conjunction with pembrolizumab in solid tumors including NSCLC (NCT03809624) (Table 1). CD137 has been shown to enhance T cell proliferation and T cell costimulatory functions and is therefore thought to act synergistically with ICI [45].

2.4. Small Cell Lung Cancer Targeting

Targeted therapy in small cell lung cancer (SCLC) has been disappointing in comparison to its NSCLC counterpart, but researchers have made substantial efforts to bridge this gap including an investigation into bispecific antibodies. In preclinical studies, DLL3 (delta-like ligand three) was found in high concentrations in SCLC tumor cells and was later found to be essential to tumorigenesis, prompting interest as a treatment target [46,47]. As rovalpituzumab tesirine was unsuccessful as an antibody–drug conjugate targeting DLL3 [48], researchers evaluated bispecific molecules that target DLL3, specifically, a bispecific T cell engager [BiTE] as an alternative (AMG757) [49]. Preclinical data on the novel agent AMG757 is promising with complete responses in both PDX models and orthotopic models due to successful T cell activation and subsequent T cell-induced tumor lysis [49]. Additionally, no treatment-related adverse events were reported with the drug [49]. In a phase I trial of this drug, now tarlatamab, 107 heavily pretreated patients with SCLC had an ORR of 23.4% [95% CI, 15.7 to 32.5] and median progression-free survival of 3.7 months [95% CI, 2.1 to 5.4] and 13.2 months [95% CI, 10.5 to not reached] [50]. Notable side effects include cytokine release syndrome and CNS toxicity [50]. This prompted a phase II trial which is currently ongoing (NCT05060016) (Table 1).

3. Antibody-Drug Conjugates

To combat the toxicity of traditional chemotherapy agents, scientists started utilizing antibodies to preferentially deliver toxic chemotherapy directly to tumor cells, so-called antibody-drug conjugates (ADCs). ADCs are monoclonal antibodies targeted to a relevant tumor antigen and linked to a chemotherapy “payload” [51]. These agents were first utilized in human clinical trials in 1983 with the ADC, anti-carcinoembryonic antigen–antibody–vindesine conjugate [51]. However, the widespread success of these agents was not achieved until the early 21st century after FDA approval of gemtuzumab ozogamicin in 2000 for CD33-positive acute myeloid leukemia (later withdrawn in 2010, but reapproved by the FDA in 2017), brentuximab vedotin in 2011 for relapsed/refractory Hodgkin’s lymphoma, and tratuzumab emtansine in 2013 for HER2 positive breast cancer [52,53,54,55]. This success fueled investigation into other targets and malignancies, including lung cancer.

3.1. HER2 Targeting

Due to the success of trastuzumab emtansine [TDM1] in HER2-positive breast cancer, there were similar hopes regarding its implementation into lung cancer, but this was more complicated with the variability in HER-2 evaluation. Currently, HER2 can be measured by protein expression [IHC], amplification [FISH cytology], and next-generation sequencing for mutations, but the results can be conflicting [56]. Initial studies of trastuzamab, a monoclonal antibody binding to HER2, alone in HER2 lung cancer (measured HER2 expression by IHC) were disappointing, but the combination with chemotherapy appeared more promising [57,58,59] (Figure 1). When trastuzumab was linked with ematansine, an antimicrotubule agent, forming an ADC, a phase II study (22 patients with HER2 exon 20 mutations) found an ORR of 38.1% [90% confidence interval, 23.0–55.9%] and median PFS and median OS of 2.8 and 8.1 months [59] (Table 2). An additional phase II trial of trastuzumab ematansine with several cohorts across different disease types that specifically analyzed NSCLC patients (N = 18 patients) with HER2 mutations by NGS rather than overexpression by IHC found ORR was 44% (95% CI, 22% to 69%) and median PFS was 5 months [95% CI, 3 to 9 months] [60] (NCT02675829).
Additional formulations of ADCs with trastuzumab were created, most notably trastuzumab deruxtecan (topoisomerase I inhibitor) in which the payload is membrane-permeable, thereby facilitating cell death in nearby cells regardless of HER2 status, the so-called “cytotoxic bystander effect” [61]. This was translated into clinical practice in a phase I trial of 60 patients with HER2 expressing non-breast/GI or HER2 mutant solid tumors with an ORR of 28.3%, but, surprisingly, ORR was 72.7%, and median PFS was 11.3 [95% CI, 8.1–14.3] months in the HER2 mutant NSCLC patients [62]. Further, in DESTINY-Lung01, a phase II study of 91 patients with previously treated HER2-mutated NSCLC, ORR was 55% [95% CI 44–64], and PFS was 8.2 [95% CI 6.0–11.9] [50] (NCT03505710) (Table 2). Significant adverse events included interstitial lung disease which occurred in 26% of the patients and was fatal in two patients [63]. DESTINY-Lung04, a phase III trial of trastuzumab deruxtecan in the first line in unresectable or metastatic NSCLC with HER2 exon 19 or 20 mutations is ongoing (NCT0504879) and will potentially provide additional insight into this line of treatment.

3.2. HER3 Targeting

Along the same line as HER2-targeted therapy in NSCLC, investigators also sought to use ADCs in HER3 targeting. HER3 expression, while common across other solid tumors, is especially relevant in lung cancer as it is expressed in 83% of NSCLC with the highest expression in EGFR-mutated NSCLC and is correlated with progression and poor relapse-free survival [64,65]. Deruxtecan was once again utilized as the drug conjugate in combination with a HER3 antibody in the novel agent, patritumab deruxtecan [66] (Figure 1). Initial data in a phase I trial of 57 patients revealed an ORR of 39% [95% CI, 26.0–52.4], and a median PFS of 8.2 [95% CI, 4.4–8.3] months [66]. The HERTHENA-Lung02 trial, a phase 3 trial of patritumab deruxtecan vs. platinum-based chemotherapy in EGFR-mutated NSCLC after EGFR TKI failure is enrolling at present (NCT05338970) (Table 2). From analysis of data from previous trials and future studies of HER3, we may be able to identify a biomarker of treatment responsiveness in addition to just HER3 expression which could help guide clinicians in the future.

3.3. TROP2 Targeting

As modifications and formulations of HER2/HER3 targeting are ongoing, new targets are also examined in this clinical space. Like HER, trophoblast cell surface antigen 2 (TROP-2), a transmembrane glycoprotein, is common among epithelial cancers including NSCLC [67]. Additionally, a large meta-analysis of 16 studies with 2569 patients found the overexpression of TROP-2 was associated with poor prognosis including overall survival with HR 1.896 [95% CI 1.599–2.247] and disease-free survival (DFS) 2.336 [95% CI 1.596–3.419] [68]. Therefore, TROP-2 was evaluated in clinical trials; the first successful use was a TROP-2 antibody conjugated to topoisomerase I inhibitor, sacituzumab govitecan, in metastatic triple-negative breast cancer with significantly improved PFS and OS compared to single-agent chemotherapy (physician choice of eribulin, vinorelbine, capecitabine, or gemcitabine) in the phase III ASCENT trial [69].
In pre-clinical studies of lung cancer, high TROP-2 expression was associated with lymph node metastasis and high histological grade; in cell lines, its overexpression increased cell proliferation and invasion [70]. This prompted further evaluation into TROP-2-based therapy in the lung along with other solid tumors with TROPION-PanTumor01 [71]. In a phase I trial of ADC, datopotamab deruxtecan (which targets TROP2) found ORR using a dose of 8 mg/kg, 25% (20/80); 6 mg/kg, 21% (8/39); and 4 mg/kg, 23% (9/40) with increased treatment-associated adverse events in the 8 mg dose group, including pneumonitis at 15% compared to 2% at the lower doses [71] [Figure 1]. TROPION-Lung01, a phase III trial to evaluate datopotamab deruxtecan vs. docetaxel in patients with advanced/metastatic NSCLC without an actionable mutation is ongoing (NCT04656652) (Table 2). Other investigations into this drug seek to evaluate its effectiveness in the NSCLC population with actionable mutations after receiving platinum-based chemotherapy with a phase 2 trial TROPION-Lung05 (NCT04484142). These ongoing trials will hopefully answer some of the questions regarding TROP-2 targeting and determine where it will fall in the hierarchy of treatment.

3.4. C-Met Targeting

As there are limited treatment options after EGFR TKI resistance in the EGFR-mutated NSCLC population, there has been a focus on the development of therapies aimed at this population. The trial of Teliso V, an ADC of a c-met antibody and monomethyl auristatin E (microtubule inhibitor), investigated the effectiveness of the drug in several different populations of NSCLC patients to determine which cohort has the most optimal response [72] (Figure 1). Initial results presented at ASCO 2022 reported an ORR of 36.5% in the non-squamous EGFR wild-type cohort but notably 52.2% ORR in the c-Met high group and 24.1% in the c-met intermediate group [73]. Surprising, both the non-squamous EGFR-mutant NSCLC group and the squamous NSCLC had a less robust response regardless of c-Met expression with ORR ranging from 11.1% to 16.7%, which prompted the closure of these cohorts/cessation of enrollment per study protocol [73]. Teliso V does appear to be well tolerated, with the most common adverse side effects being peripheral neuropathy (25%), nausea (22%), and hypoalbuminemia (20.6%) [73]. The phase 2 trial in patients with previously treated c-Met+ NSCLC is in progress (NCT03539536) (Table 2). The combination with EGFR TKIs has been promising for combating EGFR resistance; a phase 1/1b study of osimertinib therapy in combination with Teliso-V showed ORR of 50% in heavily treated, c-MET-high patients after the failure of Osimertinib monotherapy [74]. In addition, a study of erlotinib plus Teliso-V therapy had a PFS of 5.9 months, an ORR of 32.1%, and a DCR of 85.7% in patients previously treated with EGFR TKIs including c-MET-high-expressed patients [75].

3.5. AXL Targeting

In preclinical evaluations, Axl, a receptor tyrosine kinase that is part of the TAM (TYRO3, AXL, and MER) family, was shown to have several roles in cancer development including cell growth, adhesion, survival, and proliferation [76,77]. In addition, AXL is associated with higher-grade cancers, treatment resistance, and overall poor prognosis [76,78]. It is of no surprise that it has been a treatment target in many solid and hematologic malignancies [76]. In NSCLC, the activation of AXL kinase induced resistance to EGFR TKIs (erlotinib in this study), and inhibiting AXL functioning restored TKI sensitivity [79]. Enapotamab vedotin is an AXL-specific ADC that demonstrated proven activity in NSCLC in the preclinical setting [80]. The drug was evaluated across multiple tumor types in a phase I trial, but the clinical response was disappointing; therefore, the development of the drug was halted [81,82] (Table 2). Additionally, a novel ADC targeting AXL with a pyrrolobenzodiazepine cytotoxin, called Mipasetamab uzoptirine, showed promise in preclinical evaluation and is currently in a phase I clinical trial (NCT05389462) [83] (Table 2).

3.6. Emerging ADCs

After the success in the abovementioned ADCs, researchers are evaluating other combinations and molecules that may have similar or improved efficacy. Nectin-4 (cell adhesion molecule), tissue factor, and CEACAM5 (carcinoembryonic antigen-related cell adhesion molecule 5) were investigated in phase I trials in solid tumors with promising results and are now in phase II/III trials [84,85,86] (Table 2). Other molecules have been less successful in NSCLC, such as mesothelin, NaPi2b (sodium-dependent phosphate transporter), PTK7 (protein tyrosine kinase 7), and folate receptor alpha, and were not advanced past phase I [87,88,89,90] (Table 2).

4. Conclusions

Over the last two decades, the landscape of NSCLC treatment has drastically changed from the days of platinum doublet chemotherapy. Treatment options are now numerous with a significant proportion of targeted agents (NCCN NSCLC guidelines). However, responses to treatment are not forever. As is the nature of malignancy, cancer cells often develop mechanisms and alternative pathways to circumvent treatment-induced cell death. The development of ‘multi-drug resistance’ [MDR] is one postulated mechanism by which metastatic disease becomes refractory to treatment [91]. Cancer cells can develop resistance through a multitude of processes including repairing DNA damage, preventing treatment-induced apoptosis, and many others [92]. Even with treatments that target a known resistance pattern, an additional genetic mutation can render the treatment obsolete or ineffective, notably demonstrated by the acquired EGFR C797S mutation in NSCLC with preexisting EGFR T790M mutation [93]. For these reasons, investigators had to design innovative drugs that could overcome these problems.
The idea of preferentially delivering a chemotherapy agent to a cancerous cell via an antibody, antibody–drug conjugates, was one such innovative design. Another aspect of scientific breakthroughs are bispecific antibodies with the thought that targeting multiple tumor markers at the same time or bringing cytotoxic T cells in close proximity to the tumor cells could overcome known resistance. On the cumulative evaluation of these ADCs and bispecific antibodies, it does appear that they are more effective when used in a ‘precision medicine’-type approach focusing on a specific population most likely to benefit (with a specific mutation, biomarker, expression) rather than the generalized all patients/all tumor types approach utilized in typical chemotherapy. On the other hand, the pre-screening requirements of tissue biopsies (if even possible) and molecular testing substantially delay treatment initiation, so a universal agent with efficacy in all NSCLC patients could be beneficial.
One potential concern of these new agents is longevity and long-term side effects. Given the somewhat recent discovery and implementation, there is no significant follow-up data. Additionally, sequencing the novel agents with our current FDA-approved agents may lead to unforeseen complications, as was seen with the higher rates of pneumonitis with EGFR TKIs after immune checkpoint inhibitors [94]. Moreover, these ADCs/bispecifics are attempting to replace or at least be utilized as long-term maintenance therapy, like TKIs, but this is potentially problematic as the ‘payload’ (conjugated drug) is a chemotherapy agent, which are traditionally used in shorter intervals. The toxicity of the payload is influenced by several factors, including the drug–antibody ratio, the type of linker (cleavable vs. non-cleavable), and if the target is expressed commonly on adjacent normal tissue, among others [95,96].
Treatment penetration into the blood–brain barrier varies substantially by drug and has been an important factor in the disease control of NSCLC. We lack substantial data on these new drugs to determine the blood–brain barrier permeability, but a high concentration in the CSF might not be required. In prior studies, intravenous pembrolizumab demonstrated efficacy in the brain despite low levels in the CSF [97,98,99]. There is still much to discover about bispecific antibodies and antibody–drug conjugates in the treatment of lung cancer, but we can conclude that these treatment modalities are here to stay.

Author Contributions

A.R.: original drafting, R.P.: review and editing, A.M.: review and editing; E.M.: review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

EM has the following disclosures: grant/research support from Merck, & Co., Inc.; consultant for Bristol Myers Squibb, Daiichi Sankyo, Eli Lilly and Company, Genentech/Roche, Janssen Scientific Affairs, LLC, and Sanofi; on the speakers bureau for AstraZeneca, Eli Lilly and Company, Merck & Co., Inc., and Takeda Pharmaceuticals.

References

  1. Mattiuzzi, C.; Lippi, G. Current Cancer Epidemiology. J. Epidemiol. Glob. Health 2019, 9, 217–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Thai, A.A.; Solomon, B.J.; Sequist, L.V.; Gainor, J.F.; Heist, R.S. Lung cancer. Lancet 2021, 398, 535–554. [Google Scholar] [CrossRef] [PubMed]
  3. Kris, M.G.; Johnson, B.E.; Berry, L.D.; Kwiatkowski, D.J.; Iafrate, A.J.; Wistuba, I.I.; Varella-Garcia, M.; Franklin, W.A.; Aronson, S.L.; Su, P.-F.; et al. Using Multiplexed Assays of Oncogenic Drivers in Lung Cancers to Select Targeted Drugs. JAMA 2014, 311, 1998–2006. [Google Scholar] [CrossRef] [Green Version]
  4. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [Green Version]
  5. Herbst, R.S.; Giaccone, G.; de Marinis, F.; Reinmuth, N.; Vergnenegre, A.; Barrios, C.H.; Morise, M.; Felip, E.; Andric, Z.; Geater, S.; et al. Atezolizumab for First-Line Treatment of PD-L1–Selected Patients with NSCLC. N. Engl. J. Med. 2020, 383, 1328–1339. [Google Scholar] [CrossRef]
  6. Sezer, A.; Kilickap, S.; Gümüş, M.; Bondarenko, I.; Özgüroğlu, M.; Gogishvili, M.; Turk, H.M.; Cicin, I.; Bentsion, D.; Gladkov, O.; et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: A multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet 2021, 397, 592–604. [Google Scholar] [CrossRef]
  7. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.-W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef]
  8. Rosell, R.; Moran, T.; Queralt, C.; Porta, R.; Cardenal, F.; Camps, C.; Majem, M.; Lopez-Vivanco, G.; Isla, D.; Provencio, M.; et al. Screening for Epidermal Growth Factor Receptor Mutations in Lung Cancer. N. Engl. J. Med. 2009, 361, 958–967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Paez, J.G.; Janne, P.A.; Lee, J.C.; Tracy, S.; Greulich, H.; Gabriel, S.; Herman, P.; Kaye, F.J.; Lindeman, N.; Boggon, T.J.; et al. EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science 2004, 304, 1497–1500. [Google Scholar] [CrossRef] [Green Version]
  10. Lynch, T.J.; Bell, D.W.; Sordella, R.; Gurubhagavatula, S.; Okimoto, R.A.; Brannigan, B.W.; Harris, P.L.; Haserlat, S.M.; Supko, J.G.; Haluska, F.G.; et al. Activating mutations in the epidermal growth factor receptor underlying respon-siveness of non–small-cell lung cancer to gefitinib. N. Engl. J. Med. 2004, 350, 2129–2139. [Google Scholar] [CrossRef]
  11. Maemondo, M.; Inoue, A.; Kobayashi, K.; Sugawara, S.; Oizumi, S.; Isobe, H.; Gemma, A.; Harada, M.; Yoshizawa, H.; Kinoshita, I.; et al. Gefitinib or Chemotherapy for Non–Small-Cell Lung Cancer with Mutated EGFR. N. Engl. J. Med. 2010, 362, 2380–2388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Mok, T.S.; Wu, Y.-L.; Thongprasert, S.; Yang, C.-H.; Chu, D.-T.; Saijo, N.; Sunpaweravong, P.; Han, B.; Margono, B.; Ichinose, Y.; et al. Gefitinib or Carboplatin–Paclitaxel in Pulmonary Adenocarcinoma. N. Engl. J. Med. 2009, 361, 947–957. [Google Scholar] [CrossRef]
  13. Wu, Y.L.; Zhou, C.; Liam, C.K.; Wu, G.; Liu, X.; Zhong, Z.; Lu, S.; Cheng, Y.; Han, B.; Chen, L.; et al. First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR muta-tion-positive non-small-cell lung cancer: Analyses from the phase III, randomized, open-label, ENSURE study. Ann. Oncol. 2015, 26, 1883–1889. [Google Scholar] [CrossRef] [PubMed]
  14. Rosell, R.; Carcereny, E.; Gervais, R.; Vergnenegre, A.; Massuti, B.; Felip, E.; Palmero, R.; Garcia-Gomez, R.; Pallares, C.; Sanchez, J.M.; et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012, 13, 239–246. [Google Scholar] [CrossRef] [PubMed]
  15. Zhou, C.; Wu, Y.-L.; Chen, G.; Feng, J.; Liu, X.-Q.; Wang, C.; Zhang, S.; Wang, J.; Zhou, S.; Ren, S.; et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): A multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011, 12, 735–742. [Google Scholar] [CrossRef]
  16. Yu, H.A.; Arcila, M.E.; Rekhtman, N.; Sima, C.S.; Zakowski, M.F.; Pao, W.; Kris, M.G.; Miller, V.A.; Ladanyi, M.; Riely, G.J. Analysis of Tumor Specimens at the Time of Acquired Resistance to EGFR-TKI Therapy in 155 Patients with EGFR-Mutant Lung Cancers Mechanisms of Acquired Resistance to EGFR-TKI Therapy. Clin. Cancer Res. 2013, 19, 2240–2247. [Google Scholar] [CrossRef] [Green Version]
  17. Soria, J.-C.; Ohe, Y.; Vansteenkiste, J.; Reungwetwattana, T.; Chewaskulyong, B.; Lee, K.H.; Dechaphunkul, A.; Imamura, F.; Nogami, N.; Kurata, T.; et al. Osimertinib in Untreated EGFR-Mutated Advanced Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 113–125. [Google Scholar] [CrossRef]
  18. de Langen, A.J.; Johnson, M.L.; Mazieres, J.; Dingemans, A.-M.C.; Mountzios, G.; Pless, M.; Wolf, J.; Schuler, M.; Lena, H.; Skoulidis, F.; et al. Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRASG12C mutation: A randomised, open-label, phase 3 trial. Lancet 2023, 401, 733–746. [Google Scholar] [CrossRef]
  19. Shaw, A.T.; Bauer, T.M.; de Marinis, F.; Felip, E.; Goto, Y.; Liu, G.; Mazieres, J.; Kim, D.-W.; Mok, T.; Polli, A.; et al. First-Line Lorlatinib or Crizotinib in Advanced ALK-Positive Lung Cancer. N. Engl. J. Med. 2020, 383, 2018–2029. [Google Scholar] [CrossRef]
  20. Wolf, J.; Seto, T.; Han, J.-Y.; Reguart, N.; Garon, E.B.; Groen, H.J.; Tan, D.S.; Hida, T.; de Jonge, M.; Orlov, S.V.; et al. Capmatinib in MET Exon 14–Mutated or MET-Amplified Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 944–957. [Google Scholar] [CrossRef]
  21. Drilon, A.; Rekhtman, N.; Arcila, M.; Wang, L.; Ni, A.; Albano, M.; Van Voorthuysen, M.; Somwar, R.; Smith, R.S.; Montecalvo, J.; et al. A phase 2 single arm trial of cabozantinib in patients with advanced RET-rearranged lung cancers. Lancet Oncol. 2016, 17, 1653. [Google Scholar] [CrossRef] [Green Version]
  22. Drilon, A.; Siena, S.; Ou, S.H.; Patel, M.; Ahn, M.J.; Lee, J.; Bauer, T.M.; Farago, A.F.; Wheler, J.J.; Liu, S.V.; et al. Safety and Antitumor Activity of the Multitargeted Pan-TRK, ROS1, and ALK Inhibitor Entrectinib: Combined Results from Two Phase I Trials (ALKA-372-001 and STARTRK-1) Entrectinib in NTRK-, ROS1-, or ALK-Rearranged Cancers. Cancer Discov. 2017, 7, 400–409. [Google Scholar] [CrossRef] [Green Version]
  23. Mazières, J.; Zalcman, G.; Crinò, L.; Biondani, P.; Barlesi, F.; Filleron, T.; Dingemans, A.-M.C.; Léna, H.; Monnet, I.; Rothschild, S.I.; et al. Crizotinib Therapy for Advanced Lung Adenocarcinoma and a ROS1 Rearrangement: Results From the EUROS1 Cohort. J. Clin. Oncol. 2015, 33, 992–999. [Google Scholar] [CrossRef]
  24. Riudavets, M.; Sullivan, I.; Abdayem, P.; Planchard, D. Targeting HER2 in non-small-cell lung cancer (NSCLC): A glimpse of hope? An updated review on therapeutic strategies in NSCLC harbouring HER2 alterations. ESMO Open 2021, 6, 100260. [Google Scholar] [CrossRef] [PubMed]
  25. Drilon, A.; Laetsch, T.W.; Kummar, S.; Dubois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.S.; et al. Efficacy of Larotrectinib in TRK Fusion–Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef] [PubMed]
  26. Planchard, D.; Kim, T.M.; Mazieres, J.; Quoix, E.; Riely, G.; Barlesi, F.; Souquet, P.-J.; Smit, E.F.; Groen, H.J.M.; Kelly, R.J.; et al. Dabrafenib in patients with BRAFV600E-positive advanced non-small-cell lung cancer: A single-arm, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 642–650. [Google Scholar] [CrossRef] [Green Version]
  27. Cooper, A.J.; Sequist, L.V.; Lin, J.J. Third-generation EGFR and ALK inhibitors: Mechanisms of resistance and management. Nat. Rev. Clin. Oncol. 2022, 19, 499–514. [Google Scholar] [CrossRef] [PubMed]
  28. Staerz, U.D.; Kanagawa, O.; Bevan, M.J. Hybrid antibodies can target sites for attack by T cells. Nature 1985, 314, 628–631. [Google Scholar] [CrossRef]
  29. Thakur, A.; Huang, M.; Lum, L.G. Bispecific antibody based therapeutics: Strengths and challenges. Blood Rev. 2018, 32, 339–347. [Google Scholar] [CrossRef] [PubMed]
  30. Hartmann, F.; Renner, C.; Jung, W.; Deisting, C.; Juwana, M.; Eichentopf, B.; Kloft, M.; Pfreundschuh, M. Treatment of refractory Hodgkin’s disease with an anti-CD16/CD30 bispecific an-tibody. Blood 1997, 89, 2042–2047. [Google Scholar] [CrossRef]
  31. Chames, P.; Baty, D. Bispecific antibodies for cancer therapy: The light at the end of the tunnel? mAbs 2009, 1, 539–547. [Google Scholar] [CrossRef] [Green Version]
  32. Robichaux, J.P.; Elamin, Y.Y.; Tan, Z.; Carter, B.W.; Zhang, S.; Liu, S.; Li, S.; Chen, T.; Poteete, A.; Estrada-Bernal, A.; et al. Mechanisms and clinical activity of an EGFR and HER2 exon 20–selective kinase inhibitor in non–small cell lung cancer. Nat. Med. 2018, 24, 638–646. [Google Scholar] [CrossRef] [PubMed]
  33. Wu, J.Y.; Yu, C.J.; Shih, J.Y. Effectiveness of treatments for advanced non–small-cell lung cancer with exon 20 insertion epi-dermal growth factor receptor mutations. Clin. Lung Cancer 2019, 20, e620–e630. [Google Scholar] [CrossRef] [PubMed]
  34. Moores, S.L.; Chiu, M.L.; Bushey, B.S.; Chevalier, K.; Luistro, L.; Dorn, K.; Brezski, R.J.; Haytko, P.; Kelly, T.; Wu, S.-J.; et al. A Novel Bispecific Antibody Targeting EGFR and cMet Is Effective against EGFR Inhibitor–Resistant Lung Tumors. Cancer Res. 2016, 76, 3942–3953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Park, K.; Haura, E.B.; Leighl, N.B.; Mitchell, P.; Shu, C.A.; Girard, N.; Viteri, S.; Han, J.-Y.; Kim, S.-W.; Lee, C.K.; et al. Amivantamab in EGFR Exon 20 Insertion–Mutated Non–Small-Cell Lung Cancer Progressing on Platinum Chemotherapy: Initial Results From the CHRYSALIS Phase I Study. J. Clin. Oncol. 2021, 39, 3391–3402. [Google Scholar] [CrossRef]
  36. Cho, B.C.; Felip, E.; Hayashi, H.; Thomas, M.; Lu, S.; Besse, B.; Sun, T.; Martinez, M.; Sethi, S.N.; Shreeve, S.M.; et al. MARIPOSA: Phase 3 study of first-line amivantamab + lazertinib versus osimertinib in EGFR-mutant non-small-cell lung cancer. Futur. Oncol. 2022, 18, 639–647. [Google Scholar] [CrossRef]
  37. Geuijen, C.A.W.; De Nardis, C.; Maussang, D.; Rovers, E.; Gallenne, T.; Hendriks, L.J.A.; Visser, T.; Nijhuis, R.; Logtenberg, T.; de Kruif, J.; et al. Unbiased Combinatorial Screening Identifies a Bispecific IgG1 that Potently Inhibits HER3 Signaling via HER2-Guided Ligand Blockade. Cancer Cell 2018, 33, 922–936. [Google Scholar] [CrossRef] [Green Version]
  38. Schram, A.M.; Odintsov, I.; Espinosa-Cotton, M.; Khodos, I.; Sisso, W.J.; Mattar, M.S.; Lui, A.J.; Vojnic, M.; Shameem, S.H.; Chauhan, T.; et al. Zenocutuzumab, a HER2xHER3 Bispecific Antibody, Is Effective Therapy for Tumors Driven by NRG1 Gene Rearrangements. Cancer Discov. 2022, 12, 1233–1247. [Google Scholar] [CrossRef]
  39. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined nivolumab and ipilimumab or monotherapy in untreated mela-noma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [Green Version]
  40. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef] [Green Version]
  41. Sheng, I.Y.; Ornstein, M.C. Ipilimumab and Nivolumab as First-Line Treatment of Patients with Renal Cell Carcinoma: The Evidence to Date. Cancer Manag. Res. 2020, 12, 4871–4881. [Google Scholar] [CrossRef]
  42. Ascierto, P.A.; Del Vecchio, M.; Robert, C.; Mackiewicz, A.; Chiarion-Sileni, V.; Arance, A.; Lebbé, C.; Bastholt, L.; Hamid, O.; Rutkowski, P.; et al. Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: A randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2017, 18, 611–622. [Google Scholar] [CrossRef]
  43. Dovedi, S.J.; Elder, M.J.; Yang, C.; Sitnikova, S.I.; Irving, L.; Hansen, A.; Hair, J.; Jones, D.C.; Hasani, S.; Wang, B.; et al. Design and Efficacy of a Monovalent Bispecific PD-1/CTLA4 Antibody That Enhances CTLA4 Blockade on PD-1+ Activated T CellsModulated CTLA4 Inhibition via Preferential Binding to PD-1. Cancer Discov. 2021, 11, 1100–1117. [Google Scholar] [CrossRef]
  44. Ahn, M.-J.; Kim, S.-W.; Costa, E.C.; Rodríguez, L.; Oliveira, J.; Molla, M.I.; Majem, M.; Costa, L.; Su, W.-C.; Lee, K.; et al. LBA56 MEDI5752 or pembrolizumab (P) plus carboplatin/pemetrexed (CP) in treatment-naïve (1L) non-small cell lung cancer (NSCLC): A phase Ib/II trial. Ann. Oncol. 2022, 33, S1423. [Google Scholar] [CrossRef]
  45. Stoll, A.; Bruns, H.; Fuchs, M.; Völkl, S.; Nimmerjahn, F.; Kunz, M.; Peipp, M.; Mackensen, A.; Mougiakakos, D. CD137 (4-1BB) stimulation leads to metabolic and functional reprogramming of human monocytes/macrophages enhancing their tumoricidal activity. Leukemia 2021, 35, 3482–3496. [Google Scholar] [CrossRef]
  46. Owen, D.H.; Giffin, M.J.; Bailis, J.M.; Smit, M.-A.D.; Carbone, D.P.; He, K. DLL3: An emerging target in small cell lung cancer. J. Hematol. Oncol. 2019, 12, 1–8. [Google Scholar] [CrossRef] [Green Version]
  47. Sabari, J.K.; Lok, B.H.; Laird, J.H.; Poirier, J.T.; Rudin, C.M. Unravelling the biology of SCLC: Implications for therapy. Nat. Rev. Clin. Oncol. 2017, 14, 549–561. [Google Scholar] [CrossRef]
  48. Phase 3 Trial of Rova-T as Second-Line Therapy for Advanced Small-Cell Lung Cancer (TAHOE Study) Halted; AbbVie: Chicago, IL, USA, 2018.
  49. Giffin, M.J.; Cooke, K.; Lobenhofer, E.K.; Estrada, J.C.; Zhan, J.; Deegen, P.; Thomas, M.; Murawsky, C.M.; Werner, J.; Liu, S.; et al. AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer. Clin. Cancer Res. 2021, 27, 1526–1537. [Google Scholar] [CrossRef] [PubMed]
  50. Paz-Ares, L.; Champiat, S.; Lai, W.V.; Izumi, H.; Govindan, R.; Boyer, M.; Hummel, H.-D.; Borghaei, H.; Johnson, M.L.; Steeghs, N.; et al. Tarlatamab, a First-in-Class DLL3-Targeted Bispecific T-Cell Engager, in Recurrent Small-Cell Lung Cancer: An Open-Label, Phase I Study. J. Clin. Oncol. 2023, 41, 2893–2903. [Google Scholar] [CrossRef] [PubMed]
  51. Ford, C.H.; E Newman, C.; Johnson, J.R.; Woodhouse, C.S.; A Reeder, T.; Rowland, G.F.; Simmonds, R.G. Localisation and toxicity study of a vindesine-anti-CEA conjugate in patients with advanced cancer. Br. J. Cancer 1983, 47, 35–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Tsuchikama, K.; An, Z. Antibody-drug conjugates: Recent advances in conjugation and linker chemistries. Protein Cell 2016, 9, 33–46. [Google Scholar] [CrossRef] [Green Version]
  53. Sievers, E.L.; Larson, R.A.; Stadtmauer, E.A.; Estey, E.; Löwenberg, B.; Dombret, H.; Karanes, C.; Theobald, M.; Bennett, J.M.; Sherman, M.L.; et al. Efficacy and Safety of Gemtuzumab Ozogamicin in Patients with CD33-Positive Acute Myeloid Leukemia in First Relapse. J. Clin. Oncol. 2001, 19, 3244–3254. [Google Scholar] [CrossRef] [PubMed]
  54. Younes, A.; Bartlett, N.L.; Leonard, J.P.; Kennedy, D.A.; Lynch, C.M.; Sievers, E.L.; Forero-Torres, A. Brentuximab Vedotin (SGN-35) for Relapsed CD30-Positive Lymphomas. N. Engl. J. Med. 2010, 363, 1812–1821. [Google Scholar] [CrossRef] [Green Version]
  55. Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D.-Y.; Diéras, V.; Guardino, E.; et al. Trastuzumab Emtansine for HER2-Positive Advanced Breast Cancer. N. Engl. J. Med. 2012, 367, 1783–1791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Morsberger, L.; Pallavajjala, A.; Long, P.; Hardy, M.; Park, R.; Parish, R.; Nozari, A.; Zou, Y.S. HER2 amplification by next-generation sequencing to identify HER2-positive invasive breast cancer with negative HER2 immunohistochemistry. Cancer Cell Int. 2022, 22, 1–9. [Google Scholar] [CrossRef]
  57. Clamon, G.; Herndon, J.; Kern, J.; Govindan, R.; Garst, J.; Watson, D.; Green, M. Lack of trastuzumab activity in nonsmall cell lung carcinoma with overexpression of erb-B2: 39810: A phase II trial of Cancer and Leukemia Group B. Cancer Interdiscip. Int. J. Am. Cancer Soc. 2005, 103, 1670–1675. [Google Scholar]
  58. Langer, C.J.; Stephenson, P.; Thor, A.; Vangel, M.; Johnson, D.H. Trastuzumab in the Treatment of Advanced Non-Small-Cell Lung Cancer: Is There a Role? Focus on Eastern Cooperative Oncology Group Study 2598. J. Clin. Oncol. 2004, 22, 1180–1187. [Google Scholar] [CrossRef] [PubMed]
  59. Li, B.T.; Shen, R.; Buonocore, D.; Olah, Z.T.; Ni, A.; Ginsberg, M.S.; Ulaner, G.A.; Offin, M.; Feldman, D.; Hembrough, T.; et al. Ado-Trastuzumab Emtansine for Patients with HER2-Mutant Lung Cancers: Results From a Phase II Basket Trial. J. Clin. Oncol. 2018, 36, 2532–2537. [Google Scholar] [CrossRef]
  60. Iwama, E.; Zenke, Y.; Sugawara, S.; Daga, H.; Morise, M.; Yanagitani, N.; Sakamoto, T.; Murakami, H.; Kishimoto, J.; Matsumoto, S.; et al. Trastuzumab emtansine for patients with non–small cell lung cancer positive for human epidermal growth factor receptor 2 exon-20 insertion mutations. Eur. J. Cancer 2021, 162, 99–106. [Google Scholar] [CrossRef]
  61. Ogitani, Y.; Hagihara, K.; Oitate, M.; Naito, H.; Agatsuma, T. Bystander killing effect of DS-8201a, a novel anti-human epi-dermal growth factor receptor 2 antibody–drug conjugate, in tumors with human epidermal growth factor receptor 2 heter-ogeneity. Cancer Sci. 2016, 107, 1039–1046. [Google Scholar] [CrossRef]
  62. Tsurutani, J.; Iwata, H.; Krop, I.; Jänne, P.A.; Doi, T.; Takahashi, S.; Park, H.; Redfern, C.; Tamura, K.; Wise-Draper, T.M.; et al. Targeting HER2 with Trastuzumab Deruxtecan: A Dose-Expansion, Phase I Study in Multiple Advanced Solid Tumors. Cancer Discov. 2020, 10, 688–701. [Google Scholar] [CrossRef] [Green Version]
  63. Li, B.T.; Smit, E.F.; Goto, Y.; Nakagawa, K.; Udagawa, H.; Mazières, J.; Nagasaka, M.; Bazhenova, L.; Saltos, A.N.; Felip, E.; et al. Trastuzumab Deruxtecan in HER2-Mutant Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2022, 386, 241–251. [Google Scholar] [CrossRef] [PubMed]
  64. Scharpenseel, H.; Hanssen, A.; Loges, S.; Mohme, M.; Bernreuther, C.; Peine, S.; Lamszus, K.; Goy, Y.; Petersen, C.; Westphal, M.; et al. EGFR and HER3 expression in circulating tumor cells and tumor tissue from non-small cell lung cancer patients. Sci. Rep. 2019, 9, 7406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kawano, O.; Sasaki, H.; Endo, K.; Suzuki, E.; Haneda, H.; Yukiue, H.; Kobayashi, Y.; Yano, M.; Fujii, Y. ErbB3 mRNA Expression Correlated with Specific Clinicopathologic Features of Japanese Lung Cancers. J. Surg. Res. 2008, 146, 43–48. [Google Scholar] [CrossRef] [PubMed]
  66. Jänne, P.A.; Baik, C.; Su, W.-C.; Johnson, M.L.; Hayashi, H.; Nishio, M.; Kim, D.-W.; Koczywas, M.; Gold, K.A.; Steuer, C.E.; et al. Efficacy and Safety of Patritumab Deruxtecan (HER3-DXd) in EGFR Inhibitor–Resistant, EGFR-Mutated Non–Small Cell Lung Cancer. Cancer Discov. 2021, 12, 74–89. [Google Scholar] [CrossRef]
  67. Goldenberg, D.M.; Stein, R.; Sharkey, R.M. The emergence of trophoblast cell-surface antigen 2 (TROP-2) as a novel cancer target. Oncotarget 2018, 9, 28989–29006. [Google Scholar] [CrossRef] [Green Version]
  68. Zeng, P.; Chen, M.-B.; Zhou, L.-N.; Tang, M.; Liu, C.-Y.; Lu, P.-H. Impact of TROP2 expression on prognosis in solid tumors: A Systematic Review and Meta-analysis. Sci. Rep. 2016, 6, srep33658. [Google Scholar] [CrossRef] [Green Version]
  69. Bardia, A.; Hurvitz, S.A.; Tolaney, S.M.; Loirat, D.; Punie, K.; Oliveira, M.; Brufsky, A.; Sardesai, S.D.; Kalinsky, K.; Zelnak, A.B.; et al. Sacituzumab Govitecan in Metastatic Triple-Negative Breast Cancer. N. Engl. J. Med. 2021, 384, 1529–1541. [Google Scholar] [CrossRef]
  70. Li, Z.; Jiang, X.; Zhang, W. TROP2 overexpression promotes proliferation and invasion of lung adenocarcinoma cells. Biochem. Biophys. Res. Commun. 2016, 470, 197–204. [Google Scholar] [CrossRef]
  71. Meric-Bernstam, F.; Spira, A.I.; Lisberg, A.E.; Sands, J.; Yamamoto, N.; Johnson, M.L.; Yoh, K.; Garon, E.B.; Heist, R.S.; Petrich, A.; et al. TROPION-PanTumor01: Dose analysis of the TROP2-directed antibody-drug conjugate (ADC) datopotamab deruxtecan (Dato-DXd, DS-1062) for the treatment (Tx) of advanced or metastatic non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2021, 39, 9058. [Google Scholar] [CrossRef]
  72. Camidge, D.; Moiseenko, F.; Cicin, I.; Horinouchi, H.; Filippova, E.; Bar, J.; Lu, S.; Tomasini, P.; Ocampo, C.; Sullivan, D.; et al. OA15.04 Telisotuzumab Vedotin (teliso-v) Monotherapy in Patients with Previously Treated c-Met+ Advanced Non-Small Cell Lung Cancer. J. Thorac. Oncol. 2021, 16, S875. [Google Scholar] [CrossRef]
  73. Camidge, D.R.; Bar, J.; Horinouchi, H.; Goldman, J.W.; Moiseenko, F.V.; Filippova, E.; Cicin, I.; Bradbury, P.A.; Daaboul, N.; Tomasini, P.; et al. Telisotuzumab vedotin (Teliso-V) monotherapy in patients (pts) with previously treated c-Met–overexpressing (OE) advanced non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2022, 40, 9016. [Google Scholar] [CrossRef]
  74. Goldman, J.W.; Horinouchi, H.; Cho, B.C.; Tomasini, P.; Dunbar, M.; Hoffman, D.; Parikh, A.; Blot, V.; Camidge, D.R. Phase 1/1b study of telisotuzumab vedotin (Teliso-V) + osimertinib (Osi), after failure on prior Osi, in patients with advanced, c-Met overexpressing, EGFR-mutated non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2022, 40 (Suppl. 16), 9013. [Google Scholar] [CrossRef]
  75. Camidge, D.R.; Barlesi, F.; Goldman, J.W.; Morgensztern, D.; Heist, R.; Vokes, E.; Spira, A.; Angevin, E.; Su, W.-C.; Hong, D.S.; et al. Phase Ib Study of Telisotuzumab Vedotin in Combination with Erlotinib in Patients with c-Met Protein–Expressing Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2023, 41, 1105–1115. [Google Scholar] [CrossRef] [PubMed]
  76. Wium, M.; Ajayi-Smith, A.F.; Paccez, J.D.; Zerbini, L.F. The Role of the Receptor Tyrosine Kinase Axl in Carcinogenesis and Development of Therapeutic Resistance: An Overview of Molecular Mechanisms and Future Applications. Cancers 2021, 13, 1521. [Google Scholar] [CrossRef] [PubMed]
  77. Colavito, S.A. AXL as a Target in Breast Cancer Therapy. J. Oncol. 2020, 2020, 5291952. [Google Scholar] [CrossRef] [Green Version]
  78. Gjerdrum, C.; Tiron, C.; Høiby, T.; Stefansson, I.; Haugen, H.; Sandal, T.; Collett, K.; Li, S.; McCormack, E.; Gjertsen, B.T.; et al. Axl is an essential epithelial-to-mesenchymal transition-induced regulator of breast cancer metastasis and patient survival. Proc. Natl. Acad. Sci. USA 2010, 107, 1124–1129. [Google Scholar] [CrossRef] [Green Version]
  79. Zhang, Z.; Lee, J.C.; Lin, L.; Olivas, V.; Au, V.; LaFramboise, T.; Abdel-Rahman, M.; Wang, X.; Levine, A.D.; Rho, J.K.; et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet. 2012, 44, 852–860. [Google Scholar] [CrossRef]
  80. Koopman, L.A.; Terp, M.G.; Zom, G.G.; Janmaat, M.L.; Jacobsen, K.; Heuvel, E.G.-V.D.; Brandhorst, M.; Forssmann, U.; De Bree, F.; Pencheva, N.; et al. Enapotamab vedotin, an AXL-specific antibody-drug conjugate, shows preclinical antitumor activity in non-small cell lung cancer. JCI Insight 2019, 4, e128199. [Google Scholar] [CrossRef] [Green Version]
  81. Ameratunga, M.; Harvey, R.D.; Mau-Sørensen, M.; Thistlethwaite, F.; Forssmann, U.; Gupta, M.; Johannsdottir, H.; Ramirez-Andersen, T.; Bohlbro, M.L.; Losic, N.; et al. First-in-human, dose-escalation, phase (ph) I trial to evaluate safety of anti-Axl antibody-drug conjugate (ADC) enapotamab vedotin (EnaV) in solid tumors. J. Clin. Oncol. 2019, 37, 2525. [Google Scholar] [CrossRef]
  82. Genmab Announces Enapotamab Vedotin Update—Genmab A/S.Press Release. Available online: https://ir.genmab.com/news-releases/news-release-details/genmab (accessed on 28 April 2023).
  83. Zammarchi, F.; Havenith, K.E.; Chivers, S.; Hogg, P.; Bertelli, F.; Tyrer, P.; Janghra, N.; Reinert, H.W.; Hartley, J.A.; van Berkel, P.H. Preclinical Development of ADCT-601, a Novel Pyrrolobenzodiazepine Dimer-based Antibody–drug Conjugate Targeting AXL-expressing Cancers. Mol. Cancer Ther. 2022, 21, 582–593. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, Y.; Han, X.; Li, L.; Zhang, Y.; Huang, X.; Li, G.; Xu, C.; Yin, M.; Zhou, P.; Shi, F.; et al. Role of Nectin-4 protein in cancer (Review). Int. J. Oncol. 2021, 59, 93. [Google Scholar] [CrossRef] [PubMed]
  85. de Bono, J.S.; Concin, N.; Hong, D.S.; Thistlethwaite, F.C.; Machiels, J.-P.; Arkenau, H.-T.; Plummer, R.; Jones, R.H.; Nielsen, D.; Windfeld, K.; et al. Tisotumab vedotin in patients with advanced or metastatic solid tumours (InnovaTV 201): A first-in-human, multicentre, phase 1–2 trial. Lancet Oncol. 2019, 20, 383–393. [Google Scholar] [CrossRef]
  86. Johnson, M.L.; Chadjaa, M.; Yoruk, S.; Besse, B. Phase III trial comparing antibody-drug conjugate (ADC) SAR408701 with docetaxel in patients with metastatic non-squamous non-small cell lung cancer (NSQ NSCLC) failing chemotherapy and immunotherapy. J. Clin. Oncol. 2020, 38, TPS9625. [Google Scholar] [CrossRef]
  87. Shimizu, T.; Fujiwara, Y.; Yonemori, K.; Koyama, T.; Sato, J.; Tamura, K.; Shimomura, A.; Ikezawa, H.; Nomoto, M.; Furuuchi, K.; et al. First-in-Human Phase 1 Study of MORAb-202, an Antibody–Drug Conjugate Comprising Farletuzumab Linked to Eribulin Mesylate, in Patients with Folate Receptor-α–Positive Advanced Solid Tumors. Clin. Cancer Res. 2021, 27, 3905–3915. [Google Scholar] [CrossRef]
  88. Maitland, M.L.; Sachdev, J.C.; Sharma, M.R.; Moreno, V.; Boni, V.; Kummar, S.; Stringer-Reasor, E.; Lakhani, N.; Moreau, A.R.; Xuan, D.; et al. First-in-Human Study of PF-06647020 (Cofetuzumab Pelidotin), an Antibody–Drug Conjugate Targeting Protein Tyrosine Kinase 7, in Advanced Solid Tumors. Clin. Cancer Res. 2021, 27, 4511–4520. [Google Scholar] [CrossRef] [PubMed]
  89. Gerber, D.E.; Infante, J.R.; Martinez Gordon, S.; Goldberg, S.B.; Martín, M.; Felip, E.; Garcia, M.M.; Schiller, J.H.; Spigel, D.R.; Cordova, J.; et al. Phase Ia Study of Anti-NaPi2b Antibody–Drug Conjugate Lifastuzumab Vedotin DNIB0600A in Patients with Non–Small Cell Lung Cancer and Platinum-Resistant Ovarian Cancer. Clin. Cancer Res. 2020, 26, 364–372. [Google Scholar] [CrossRef] [Green Version]
  90. Hassan, R.; Blumenschein, G.R., Jr.; Moore, K.N.; Santin, A.D.; Kindler, H.L.; Nemunaitis, J.J.; Seward, S.M.; Thomas, A.; Kim, S.K.; Rajagopalan, P.; et al. First-in-Human, Multicenter, Phase I Dose-Escalation and Expansion Study of Anti-Mesothelin Antibody–Drug Conjugate Anetumab Ravtansine in Advanced or Metastatic Solid Tumors. J. Clin. Oncol. 2020, 38, 1824–1835. [Google Scholar] [CrossRef]
  91. Cui, Q.; Wang, J.-Q.; Assaraf, Y.G.; Ren, L.; Gupta, P.; Wei, L.; Ashby, C.R., Jr.; Yang, D.-H.; Chen, Z.-S. Modulating ROS to overcome multidrug resistance in cancer. Drug Resist. Updat. 2018, 41, 1824–1835. [Google Scholar] [CrossRef]
  92. Kachalaki, S.; Ebrahimi, M.; Khosroshahi, L.M.; Mohammadinejad, S.; Baradaran, B. Cancer chemoresistance; biochemical and molecular aspects: A brief overview. Eur. J. Pharm. Sci. 2016, 89, 20–30. [Google Scholar] [CrossRef]
  93. Leonetti, A.; Sharma, S.; Minari, R.; Perego, P.; Giovannetti, E.; Tiseo, M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br. J. Cancer 2019, 121, 725–737. [Google Scholar] [CrossRef]
  94. Chan, D.W.-K.; Choi, H.C.-W.; Lee, V.H.-F. Treatment-Related Adverse Events of Combination EGFR Tyrosine Kinase Inhibitor and Immune Checkpoint Inhibitor in EGFR-Mutant Advanced Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Cancers 2022, 14, 2157. [Google Scholar] [CrossRef] [PubMed]
  95. Theocharopoulos, C.; Lialios, P.-P.; Samarkos, M.; Gogas, H.; Ziogas, D.C. Antibody-Drug Conjugates: Functional Principles and Applications in Oncology and Beyond. Vaccines 2021, 9, 1111. [Google Scholar] [CrossRef] [PubMed]
  96. Tarantino, P.; Pestana, R.C.; Corti, C.; Modi, S.; Bardia, A.; Tolaney, S.M.; Cortes, J.; Soria, J.; Curigliano, G. Antibody–drug conjugates: Smart chemotherapy delivery across tumor histologies. CA A Cancer J. Clin. 2021, 72, 165–182. [Google Scholar] [CrossRef] [PubMed]
  97. Portnow, J.; Wang, D.; Blanchard, M.S.; Tran, V.; Alizadeh, D.; Starr, R.; Dodia, R.; Chiu, V.; Brito, A.; Kilpatrick, J.; et al. Systemic Anti–PD-1 Immunotherapy Results in PD-1 Blockade on T Cells in the Cerebrospinal Fluid. JAMA Oncol. 2020, 6, 1947–1951. [Google Scholar] [CrossRef] [PubMed]
  98. Portnow, J.; Synold, T.; Tran, V.; Chiu, V.; Alizadeh, D.; Wang, D.; Brito, A.; Kilpatrick, J.; McNamara, P.; Forman, S.; et al. ATIM-17. PEMBROLIZUMAB BLOCKS PD-1 ON CAR T CELLS ADMINISTERED INTRAVENTRICULARLY TO GLIOBLASTOMA PATIENTS. Neuro-Oncol. 2018, 20, vi4. [Google Scholar] [CrossRef]
  99. Goldberg, S.B.; Schalper, K.A.; Gettinger, S.N.; Mahajan, A.; Herbst, R.S.; Chiang, A.C.; Lilenbaum, R.; Wilson, F.H.; Omay, S.B.; Yu, J.B.; et al. Pembrolizumab for management of patients with NSCLC and brain metastases: Long-term results and biomarker analysis from a non-randomised, open-label, phase 2 trial. Lancet Oncol. 2020, 21, 655–663. [Google Scholar] [CrossRef]
Figure 1. Biochemical Pathways: antibody-drug conjugates and bispecific antibodies targets Designed using BioReader.
Figure 1. Biochemical Pathways: antibody-drug conjugates and bispecific antibodies targets Designed using BioReader.
Cancers 15 03162 g001
Table 1. Bispecific antibodies in lung cancer.
Table 1. Bispecific antibodies in lung cancer.
Drug Name Trial NCT
AmivantamabMARIPOSA1/Phase 3NCT04487080
AZD-9592EGRET/Phase 1NCT05647122
TarlatamabDeLLphi-301/Phase 2NCT05060016
MEDI5752Phase 1NCT03530397
ZenocutuzumabPhases 1 and 2NCT02912949
INBRX-105Phase 2NCT03809624
Trial data obtained from NIH clinicaltrials.gov.
Table 2. Antibody-Drug Conjugates in Lung Cancer.
Table 2. Antibody-Drug Conjugates in Lung Cancer.
Drug Name Trial TargetNCT
Enapotamab VedotinPhase 1AxlNCT02988817
Trastuzumab EmtansinePhase 2HER2NCT02289833
Trastuzumab DeruxtecanDESTINY-Lung01HER2NCT03505710
Patritumab DeruxtecanHERTHENA-Lung02HER3NCT05338970
Datopotamab DeruxtecanTROPION-Lung01TROP2NCT04656652
Teliso-VPhase 2c-MetNCT03539536
Mipasetamab uzoptirinePhase 1AxlNCT05389462
Enfortumab VedotinPhase 2Nectin-4NCT04225117
Tisotumab VedotinInnovaTV 201Tissue FactorNCT02001623
Farletuzumab EcteribulinPhase 1Folate receptor alphaNCT03386942
Cofetuzumab PelidotinPhase 1PTK7NCT02222922
Tusamitamab RavtansineCARMEN-LC03CEACAM5NCT04154956
Lifastuzumab VedotinPhase 1NaPi2bNCT01363947
Trial data obtained from NIH clinicaltrials.gov.
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

Reyes, A.; Pharaon, R.; Mohanty, A.; Massarelli, E. Arising Novel Agents in Lung Cancer: Are Bispecifics and ADCs the New Paradigm? Cancers 2023, 15, 3162. https://doi.org/10.3390/cancers15123162

AMA Style

Reyes A, Pharaon R, Mohanty A, Massarelli E. Arising Novel Agents in Lung Cancer: Are Bispecifics and ADCs the New Paradigm? Cancers. 2023; 15(12):3162. https://doi.org/10.3390/cancers15123162

Chicago/Turabian Style

Reyes, Amanda, Rebecca Pharaon, Atish Mohanty, and Erminia Massarelli. 2023. "Arising Novel Agents in Lung Cancer: Are Bispecifics and ADCs the New Paradigm?" Cancers 15, no. 12: 3162. https://doi.org/10.3390/cancers15123162

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