Over the last several years, several molecular aberrations have been unevenly described across cancers, although the distinct functional relevance in each biological context is not yet fully understood. Novel discoveries have led to the development of drugs tailored to the molecular profile of patients, thus increasing the likelihood of response among biomarker-selected patients. In this context, there has been a progressive redefinition of a precision medicine framework where evidence-based development and earlier approvals might now be driven by this molecular information. Innovative trial designs have greatly facilitated the evaluation and approval of new drugs in small cohorts of orphan cancers in which histology-dependent molecularly defined trials might be logistically difficult. However, accelerated approvals based on this agnostic-histology development model have brought new clinical, regulatory, and reimbursement challenges. In this article, we will highlight many of the biologic issues and clinical trial design challenges characterizing the development of tissue-agnostic compounds. Also, we will review some of the key factors involved in the development of pembrolizumab and larotrectinib, the first two drugs that have been approved by the U.S. Food and Drug Administration in an histology-agnostic manner. Because we anticipate that agnostic-histology approvals will continue to grow, we aim to provide insight into the current panorama of targeted drugs that are following this strategy and some premises to take into consideration. Clinicians and regulators should be prepared to overcome the associated potential hurdles, ensuring that uncertainties are dealt with properly and allowing new, promising agents to arrive faster to the market.

In recent years, advances in high-throughput technologies have yielded impressive insights into the molecular biology behind cancers. Novel discoveries have resulted in the development of several innovative drugs, representing a paradigm shift in the oncology early drug development panorama (1, 2). Cancer-omics have become a powerful ally for applying biomarkers and focusing clinical development in molecularly selected populations, although they have also brought another layer of complexity (3).

In consequence, we are witnessing the emergence of a new era characterized by the recognition of tumors as genetic diseases, accelerating the development timings from the molecular aberration discovery to the approval of new drugs (Fig. 1; ref. 4). Although more clinical evidence-based information is gathered, several classifications have been developed to guide clinicians to prioritize molecular biomarkers to tailor patients with the most appropriate targeted agents [e.g., European Society for Medical Oncology (ESMO) Scale for Clinical Actionability of molecular Targets (ESCAT); ref. 5]. In this context, histology-agnostic trials might represent the ideal scenario for gaining a better understanding of the functional relevance of a certain molecular aberration across multiple tumor types (6).

Figure 1.

The evolving framework of clinical trials used to support oncology drug approval. Trials have shifted from enrolling (i) unselected patients exclusively based on the type of tumor to an exquisite, appropriate selection of biomarker-defined populations either (ii) within a specific tumor type or (iii) across a variety of different cancers that share a common molecular abnormality. Consequently, phase III (ph3) clinical trials have been gradually replaced by redesigned phase I (ph1) and II (ph2) clinical trials, recently leading to accelerated and conditional approvals of new anticancer agents based on the results of phase I/II “basket” trials and large expansion cohorts of molecularly selected patients, thus redefining the traditional phase I/II/III model that previously worked with cytotoxic chemotherapies.

Figure 1.

The evolving framework of clinical trials used to support oncology drug approval. Trials have shifted from enrolling (i) unselected patients exclusively based on the type of tumor to an exquisite, appropriate selection of biomarker-defined populations either (ii) within a specific tumor type or (iii) across a variety of different cancers that share a common molecular abnormality. Consequently, phase III (ph3) clinical trials have been gradually replaced by redesigned phase I (ph1) and II (ph2) clinical trials, recently leading to accelerated and conditional approvals of new anticancer agents based on the results of phase I/II “basket” trials and large expansion cohorts of molecularly selected patients, thus redefining the traditional phase I/II/III model that previously worked with cytotoxic chemotherapies.

Close modal

In this article, we will review the clinical development of a number of compounds that were early assessed in agnostic-histology trials and which lessons were learnt from these initial studies. Moreover, we will then depict the key factors involved in the development of pembrolizumab and larotrectinib, the first drugs approved by the U.S. Food and Drug Administration (FDA) in an histology-agnostic manner, although not yet by the European Medicines Agency. Finally, we aim to provide insight into the current panorama of promising drugs that are following this strategy and its associated potential hurdles.

The biological relevance of molecular aberrations and the response to targeted agents can substantially differ depending on tumor type (7). Two of the most illustrative cases are the development of the v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) and the HER2 inhibitors.

The BRAF inhibitor (BRAFinh) vemurafenib, showed encouraging efficacy among patients with melanoma enrolled in a phase I trial (81% complete or partial response; ref. 8). Posterior identification of reactivation of the mitogen-activated protein kinase (MAPK) pathway as an escape mechanism led to the approval of an evidence-based combination strategy of BRAFinh and mitogen-activated extracellular signal-related kinase (MEK) inhibitors for BRAF mutant (BRAFmut) patients with melanoma (9). This combined approach drastically changed the outcome of this disease. However, only 5% of BRAFmut patients with colon cancer responded to BRAFinh monotherapy, translating the heterogeneous pattern of BRAF activation within this histology (10). Further preclinical insights evidenced that the BRAFinh effect could be rapidly bypassed by ERK reactivation through an EGFR-mediated activation of RAS and C-RAF (11, 12). Unfortunately, triple strategies of BRAF/MEK/EGFR inhibition have not paralleled the efficacy reached in melanoma, mainly due to the continued adaptive feedback reactivation of the MAPK signaling in the colorectal cancer context (13).

Similarly, the VE-Basket trial testing vemurafenib in BRAFmut V600 non-melanoma cancers, reported isolated responses among a myriad of tumor types (14). Non–small cell lung cancer (NSCLC) cases and patients with Erdheim-Chester disease (ECD) seemed to retain clear oncogene-addiction to BRAFmut, achieving an overall response rate (ORR) of 42% [95% confidence interval (CI), 20-67] and 43% (95% CI, 18–71), respectively. Remarkably, the results of this study led to the FDA approval for vemurafenib in ECD in 2017 (15). Furthermore, the study highlighted that BRAFinh would not act homogenously across histologies, since depending on the biological role of BRAF, distinct resistance mechanisms might develop.

Another example worth mentioning is the case of HER2 inhibitors (HER2inh). Trastuzumab, the first humanized HER2-directed monoclonal antibody (mAb), received FDA approval after demonstrating survival benefit for patients with HER2-amplified (HER2amp) metastatic breast cancer (16). In 2010, trastuzumab also showed increased overall survival (OS) from 11.1 to 13.8 months [hazard ratio (HR) for death 0.74; 95% CI, 0.60–0.91; P = 0.0046] in patients with HER2amp gastroesophageal cancer (GE) in the ToGA trial (17). The HERACLES study demonstrated that the combination of lapatinib plus trastuzumab was active in a subset of HER2amp patients with refractory colorectal cancer (5%), achieving an ORR of 30% (95% CI, 14–50; ref. 18). Noteworthy, low prevalence of HER2amp has also been reported among other miscellaneous tumor types, thus encouraging the research of novel HER2inh beyond the classical indications. ZW25, a bispecific antibody targeting ECD2–ECD4 HER2 domains, showed a disease control rate (DCR) of 33% in non-breast/GE tumors overexpressing HER2 (19). Also, trials testing different antibody–drug conjugates, such as ado-trastuzumab emtansine (T-DM1; ref. 20) and trastuzumab deruxtecan (DS-8201a; ref. 21), have confirmed clinical activity of the HER2 blockade among other HER2-expressing cancers (e.g., salivary gland).

Lately, rising evidence of mutations in HER2 (HER2mut) widened further exploration of anti-HER2 therapies (22). The MOSCATO-01/02 trials showed that the clinical benefit rate (CBR) to HER2 blockade for HER2amp patients was significantly higher (CBR 64%) compared with those HER2mut (CBR 25%; ref. 23). In the SUMMIT trial, patients with HER2/3mut across 21 tumor types were treated with neratinib, a pan-HER multikinase inhibitor. No activity was observed in the HER3mut cohort. On the contrary, among HER2mut patients (n = 124), responses were reported among NSCLC, breast, biliary, salivary and cervical cancers, with the greatest activity being seen in kinase domain missense mutations. To date, this study has provided the most comprehensive data that exist on the differential clinical actionability of HER2/3mut (24).

Several concepts were learnt from these initial trials developing BRAFinh and HER2inh. First, it became clear that the molecular status alone is not enough to predict response, as the signaling pathway might play different roles in each specific histologic context, where different mechanistic explanations could mediate resistances. Second, the results revealed that efficacy might be influenced by the class of molecular alteration (amp > mut), the involved gene (HER2 > HER3) or, the type of allele (mut in the kinase domain > frameshifts in receptor domain). Finally, these studies demonstrated that molecularly guided trials could be used to refine our biological understanding of targets, and that conventional cancer therapy should not be solely replaced by the molecular findings. Investigators should be careful when extrapolating the outcomes seen with one drug from one cancer to another, keeping in mind that significant differences exist between different histologies, and profound heterogeneity has been observed even within one same cancer type.

Microsatellite instability-high (MSI-H) tumors can have up to eight times more somatic mutations than microsatellite stable (MSS) cancers, translating into a higher presence of neoantigens able to triggering an immune response (25). Checkpoint blockade with anti-programmed death-1 (PD-1) antibodies was hypothesized to enhance immune responses in MSI-H/deficient mismatch repair (dMMR) tumors, supported by the idea that a higher mutational load was correlated with responses in melanoma (26) and NSCLC (27). All these observations provided the incentive for the development of the KEYNOTE-016 trial, which selectively investigated pembrolizumab in MSI/dMMR patients.

Patients with dMMR colorectal cancer, mismatch repair-proficient (MMR) colorectal cancer, and patients with dMMR non-colorectal cancer refractory cancers were enrolled in the KEYNOTE-016 study. The co-primary endpoints were immune-related objective response rate (irRR) and 20-week immune-related progression free survival (irPFS) rate. irRR were 40% and 71% for patients with dMMR colorectal cancer and dMMR non-colorectal cancer tumors, respectively. The median progression-free survival (mPFS) and mOS were not reached at the time of the report, and the irPFS rates were 78% for the dMMR colorectal cancer cohort and 67% for the dMMR non-colorectal cancer group. Noteworthy, the efficacy among MMR colorectal cancer was meaningless, with 0% irRR, 11% irPFS, and a mOS of 5 months (HR, 0.22; P = 0.05). Whole-exome sequencing (WES) revealed that dMMR tumors presented almost 24 times more somatic mutations compared with MMR ones (P = 0.007). As expected, higher mutation loads were associated with prolonged mPFS (P = 0.02; ref. 28).

In addition to KEYNOTE-016 (n = 58), two subsequent studies also provided robust evidence of antitumor activity of pembrolizumab and durable responses in heavily pretreated dMMR patients. The KEYNOTE-164 trial enrolled patients with MSI-H colorectal cancer who had received at least 2 prior lines of therapy (n = 61), whereas the multi-cohort KEYNOTE-158 study included patients with MSI-H non-colorectal cancer treated with at least one prior regimen (n = 19; refs. 29, 30). Furthermore, patients with PD-L1–positive tumors and MSI-H enrolled in phase I studies assessing pembrolizumab were retrospectively identified [KEYNOTE-012, n = 6 (refs. 31–33); KEYNOTE-028, n = 5 (refs. 34–38)].

On May 23, 2017, the FDA granted the accelerated approval of pembrolizumab for the treatment of adult and pediatric patients with unresectable or metastatic refractory MSI-H/dMMR tumors. The first FDA tissue-agnostic approval became a reality, based on 149 patients with MSI-H/dMMR tumors across the five uncontrolled, single-arm trials depicted in Table 1 (ref. 39). ORR was similar irrespectively of the origin (36% in colorectal cancer vs. 46% in 14 other cancer types). Despite this accelerated approval, continued approval of pembrolizumab for this indication will rely on the final results of clinical benefit in the ongoing confirmatory trials.

Table 1.

Studies that supported the agnostic-histology approval of pembrolizumab for MSI-H/dMMR solid tumors by the FDA

StudyDesignNTumorsPrior therapyMSI-H/dMMR testingDoseClinicalTrial.gov IDRef
Prospective enrollment of patients with MSI-H/dMMR tumors 
KEYNOTE-016 Investigator-initiated ph2 at 6 sites 58 Multiple ≥1 prior regimen non-CRC and ≥2 prior regimen CRC Local PCR or IHC 10 mg/kg every 2 weeks NCT01876511 (28–30) 
KEYNOTE-158 International and multicenter ph2 19 Multiple ≥1 prior regimen Local PCR (central for non-CRC rare tumors) or IHC 200 mg every 3 weeks NCT02628067  
KEYNOTE-164 International and multicenter ph2 61 CRC Prior fluoropyrimidine, oxaliplatin, irinotecan ± anti-VEGF/EGFR mAb Local PCR or IHC 200 mg every 3 weeks NCT02460198  
Retrospective identification of patients with MSI-H/dMMR tumors 
KEYNOTE-012 Multicenter ph1b Gastric, bladder, or TNBC ≥1 prior regimen Central PCR 10 mg/kg every 2 weeks NCT01848834 (31–33) 
KEYNOTE-028 Multicenter ph1 Esophageal, CRC, breast, endometrial, or biliary ≥1 prior regimen Central PCR 10 mg/kg every 2 weeks NCT02054806 (34–38) 
StudyDesignNTumorsPrior therapyMSI-H/dMMR testingDoseClinicalTrial.gov IDRef
Prospective enrollment of patients with MSI-H/dMMR tumors 
KEYNOTE-016 Investigator-initiated ph2 at 6 sites 58 Multiple ≥1 prior regimen non-CRC and ≥2 prior regimen CRC Local PCR or IHC 10 mg/kg every 2 weeks NCT01876511 (28–30) 
KEYNOTE-158 International and multicenter ph2 19 Multiple ≥1 prior regimen Local PCR (central for non-CRC rare tumors) or IHC 200 mg every 3 weeks NCT02628067  
KEYNOTE-164 International and multicenter ph2 61 CRC Prior fluoropyrimidine, oxaliplatin, irinotecan ± anti-VEGF/EGFR mAb Local PCR or IHC 200 mg every 3 weeks NCT02460198  
Retrospective identification of patients with MSI-H/dMMR tumors 
KEYNOTE-012 Multicenter ph1b Gastric, bladder, or TNBC ≥1 prior regimen Central PCR 10 mg/kg every 2 weeks NCT01848834 (31–33) 
KEYNOTE-028 Multicenter ph1 Esophageal, CRC, breast, endometrial, or biliary ≥1 prior regimen Central PCR 10 mg/kg every 2 weeks NCT02054806 (34–38) 

Abbreviations: Anti-VEGF/EGFR mAb, anti-vascular endothelial growth factor/epidermal growth factor receptor mAb; CRC, colorectal cancer; IHC, immunohistochemistry; N, number of patients enrolled; PCR, polymerase chain reaction; ph1, phase I clinical trial; ph2, phase II clinical trial; Ref, Reference; TNBC, triple-negative breast cancer.

What have we learnt from this historical approval? First, a strong biologic preclinical and clinical rationale was needed, ensuring the homogeneity of effects of this alteration among different tissues. In this case, MSI tumors were found across multiple histologies at varying frequencies (40), sharing common features known to be predictive of response to immunotherapy, such as PD-L1 expression, high tumor mutational burden (TMB) and lymphocyte T infiltration (41). Also, MSI tumors undergo immune-editing processes that provide them with genetic advantages for immune escape, most likely a common resistance trait across MSI cancers (42). Second, it was important to measure the expected efficacy with a reliable endpoint, facilitating the interpretation of results across histologies inside one trial and between studies. Noteworthy, similar ORR and OS/PFS rates were reported in at least 14 tumor types in the pooled analysis for pembrolizumab, with no differential pattern of efficacy indicating a histology qualitative effect (29). Third, a truly agnostic indication would embrace the treatment of both, adult and pediatric patients, ensuring an appropriate formulation for the younger. In fact, the biology of MSI-H/dMMR was expected to be similar in pediatric tumors (43), and there was an already established dose of pembrolizumab for pediatric Hodgkin lymphoma (KEYNOTE-051 study; ref. 44). Finally, the possibility of identifying those patients whose tumors harbor the predictive biomarker with a validated companion diagnostic is a key limitant factor for ensuring the attrition of patients. In this case, immunohistochemistry (IHC) testing or polymerase chain reaction (PCR)–based assays were commercially available (45).

Following the approval of pembrolizumab, another milestone revolutioned the agnostic-histology scene in 2017. Patients with neurotrophin tropomyosin receptor kinase (NTRK) rearranged (rear) tumor types were prospectively identified and enrolled into the adult phase I (n = 8) or “basket” phase II (n = 35), and the pediatric phase I/II (n = 12) trials evaluating larotrectinib, a first-in-class pan-TRK inhibitor (TRKinh; ref. 46). The ORR was 75% (95% CI, 61–85) among 12 different histologies, ranging in ages from 4 months to 76 years. The most common cancers were salivary gland (22%), soft tissue sarcoma (20%), infantile fibrosarcoma (13%) and thyroid (9%). Strikingly, larotrectinib demonstrated clinical benefit regardless of tumor type, NTRK gene, fusion partner or age of the patient. Also, encouraging central nervous system activity was seen in primary malignancies and metastatic lesions, with rapid and prolonged responses. On the basis the results of these 55 patients, on November 26, 2018, the FDA granted accelerated approval of larotrectinib for the treatment of adult and pediatric NTRKrear tumors (47).

Interestingly, the treatment of larotrectinib has been approved based on the presence of a NTRKrear in tumor specimens. However, unlike the approval of pembrolizumab for MSI-H/dMMR tumors, there is not an FDA-approved test for the detection of NTRKrear. Recently, updated results of efficacy with larotrectinib were presented with the expanded 122-patient integrated dataset, showing an ORR of 81% (95% CI, 72–88). Remarkably, 84% of the responding patients and 73% of all patients remained on larotrectinib or underwent surgery with curative intent (48). These highly consistent results might have fostered the approval of larotrectinib, allowing the detection of NTRKrear based on local NGS or FISH.

Pembrolizumab was the first tissue-agnostic drug approved but larotrectinib was the first drug developed with an agnostic-histology indication in mind. The truth is, gene fusions have emerged as an important class of somatic alterations, acting as oncogenic drivers in approximately 16.5% of human cancers (49). It is understandable then that these druggable fusions have focused the attention of the clinical research field.

Pooled data from phase I/II entrectinib trials showed an ORR of 57.4% (95% CI, 43.2–70.8) among NTRKrear adult patients from 19 different histopathologies, also with durable intracraneal responses (54.5%; ref. 50). In parallel, the discovery of early acquired mechanisms of resistance, has led the fast development of successful second-generation TRKinh [e.g., loxo-195 (ref. 51), repotrectinib (ref. 52) or merestinib (ref. 53)]. Moreover, debio1347, a selective FGFR1/2/3 inhibitor (FGFRinh), showed promising efficacy among patients harboring fibroblast growth factor receptor (FGFR) fusions. A DCR of 87% was reported in 5 different histologies with FGFRrear (n = 8; ref. 54). In view of these results, other “basket” trials are further developing FGFRinh in this difficult-to-find FGFRrear population (e.g., TAS-120; ref. 55). Later this year, the RET inhibitor (RETinh) loxo-292 showed an encouraging ORR of 77% (95% CI, 61–89) in RETrear evaluable tumors (77% in NSCLC and 78% in other cancers), including intracranial responses in NSCLC (56). Also, many other promising compounds are focusing their development in this small RETrear population [e.g., agerafenib (ref. 57) and BLU-667 (ref. 58)]. With the arrival of these best-in-class molecular targeted agents (MTA), the early drug development panorama is becoming increasingly busy, and what matters most, it seems more promising than ever. Table 2 summarizes the current approval status of some of these MTA.

Table 2.

Selected tyrosine kinase inhibitors under agnostic-histology clinical development

DrugTargetMolecularly selected populationFDA statusEMA statusClinicalTrial.gov ID
Debio1347 FGFR1/2/3 Ph1/2 “basket” in solid tumors with FGFR fusions. May 2018: Fast Track designation for tumors with specific FGFR alteration. November 2017: Orphan Drug designation for BTC. NCT01948297 
TAS-120 FGFR1/2/3/4 Ph1/2 “basket” in solid tumors with FGFR fusions. May 2018: Orphan Drug designation for iCCA. N/E NCT02052778 
Larotrectinib TRK Ph2 “basket” in adults with NTRKrear solid tumors (NAVIGATE study)November 2018: Accelerated Approval. August 2018: MarketingAuthorization Application submitted. NCT02576431 
  Ph1/2 “basket” in children with NTRKrear solid tumors (SCOUT study)  NCT02637687 
  Ph1 in adults with NTRKrear solid tumors (LOXO-TRK-14001 study)  NCT02122913 
Entrectinib (RXDX-101) TRK/ALK/ROS1 Ph2 “basket” in adults with NTRK/ALK/ROS1rear solid tumors (STARTRK-2 study)July 2017: Orphan Drug designation for adult/children with NTRKrear solid tumors. October 2017: Priority Medicines designation for adult/children with NTRKrear solid tumors. NCT02568267 
  Ph1/1b in children with NTRK/ALK/ROS1rear solid tumors (STARTRK-NG study)  NCT02650401 
Merestinib (LY2801653) TRK/MET Ph2 in NSCLC MET exon 14 mut or “basket” for NTRKrear solid tumors. October 2017: Orphan Drug designation for BTC. N/E NCT02920996 
Repotrectinib (TPX-0005) TRK/ALK/ROS1 Ph1/2 in NTRK/ALK/ROS1rear solid tumors (TRIDENT-1 study)June 2017: Orphan Drug designation for NTRK/ALK/ROS1rear NSCLC. N/E NCT03093116 
Loxo-195 TRK Ph1/2 in adult and children previously treated with NTRKrear cancers. May 2018: NDA accepted and granted Priority Review for adults and children with NTRKrear tumors. N/E NCT03215511 
Loxo-292 RET Ph1/2 in RETrear solid tumors and MTC (LIBRETTO-001 study)September 2018: Breakthrough designation for RETrear NSCLC and RETmut MTC. N/E NCT03157128 
Agerafenib (RXDX-105) RET/BRAF Ph1/1b in patients with advanced solid tumors. N/E N/E NCT01877811 
BLU-667 RET Ph1 in RET-altered solid tumors and MTC (ARROW study)N/E N/E NCT03037385 
DrugTargetMolecularly selected populationFDA statusEMA statusClinicalTrial.gov ID
Debio1347 FGFR1/2/3 Ph1/2 “basket” in solid tumors with FGFR fusions. May 2018: Fast Track designation for tumors with specific FGFR alteration. November 2017: Orphan Drug designation for BTC. NCT01948297 
TAS-120 FGFR1/2/3/4 Ph1/2 “basket” in solid tumors with FGFR fusions. May 2018: Orphan Drug designation for iCCA. N/E NCT02052778 
Larotrectinib TRK Ph2 “basket” in adults with NTRKrear solid tumors (NAVIGATE study)November 2018: Accelerated Approval. August 2018: MarketingAuthorization Application submitted. NCT02576431 
  Ph1/2 “basket” in children with NTRKrear solid tumors (SCOUT study)  NCT02637687 
  Ph1 in adults with NTRKrear solid tumors (LOXO-TRK-14001 study)  NCT02122913 
Entrectinib (RXDX-101) TRK/ALK/ROS1 Ph2 “basket” in adults with NTRK/ALK/ROS1rear solid tumors (STARTRK-2 study)July 2017: Orphan Drug designation for adult/children with NTRKrear solid tumors. October 2017: Priority Medicines designation for adult/children with NTRKrear solid tumors. NCT02568267 
  Ph1/1b in children with NTRK/ALK/ROS1rear solid tumors (STARTRK-NG study)  NCT02650401 
Merestinib (LY2801653) TRK/MET Ph2 in NSCLC MET exon 14 mut or “basket” for NTRKrear solid tumors. October 2017: Orphan Drug designation for BTC. N/E NCT02920996 
Repotrectinib (TPX-0005) TRK/ALK/ROS1 Ph1/2 in NTRK/ALK/ROS1rear solid tumors (TRIDENT-1 study)June 2017: Orphan Drug designation for NTRK/ALK/ROS1rear NSCLC. N/E NCT03093116 
Loxo-195 TRK Ph1/2 in adult and children previously treated with NTRKrear cancers. May 2018: NDA accepted and granted Priority Review for adults and children with NTRKrear tumors. N/E NCT03215511 
Loxo-292 RET Ph1/2 in RETrear solid tumors and MTC (LIBRETTO-001 study)September 2018: Breakthrough designation for RETrear NSCLC and RETmut MTC. N/E NCT03157128 
Agerafenib (RXDX-105) RET/BRAF Ph1/1b in patients with advanced solid tumors. N/E N/E NCT01877811 
BLU-667 RET Ph1 in RET-altered solid tumors and MTC (ARROW study)N/E N/E NCT03037385 

Abbreviations; ALK, anaplastic lymphoma kinase; BTC, biliary tract cancers; EMA, European Medicines Agency; iCCA, intrahepatic colangiocarcinoma; MTC, medullary thyroid cancer; mut, mutant; N/E, not evaluated; NDA, New Drug Application; Ph1, phase I clinical trial; Ph2, phase II clinical trial; rear, rearranged.

With the increasing evidence of immune-predictive biomarkers commonly displayed by cancers, tissue-agnostic development is focusing on the immune-checkpoint inhibitors arena (59). In light that anti–PD-1 therapies have shown efficacy across different cancers, efforts have now focused on the identification of tissue-agnostic immune-biomarkers. In reality, MSI-H might have been a surrogate marker of high TMB (60). Therefore, TMB is emerging as an independent promising biomarker for anti–PD-1/-L1 therapy, and WES methods are investigating its applicability across a full spectrum of tumor types (61).

Although there continues to be an urgent unmet medical need for patients with refractory cancers, the development of new drugs in an agnostic-histology manner will probably increase. However, this model of accelerated drug development, based on unprecedented efficacy coming from single-arm small cohorts of biomarker-selected patients will be an exception rather than a rule.

The increasing evidence of a significant myriad of genomic alterations across different cancers has highlighted that it is getting more difficult to explore rare signatures in one single histology. Tumor agnostic drug development strategies could address this myriad, although improving precision medicine in cancer will certainly require more flexibility from all the participants involved (62).

First, the clinical framework for drug development will need to adapt accordingly. Sophisticated master protocols, including “umbrella,” “platform,” and “basket” trials, have been designed to answer multiple questions in less time (63). Two examples of these ambitious approaches are the NCI-MATCH trial, which pretends to enroll more than 1,000 all-comer patients in 25 targeted arms (64), or the Basket of Baskets trial, the largest personalized cancer medicine trial in Europe (65). Figure 2 illustrates the integration of these innovative designs with early translational strategies, serving as platforms to advance research (66). Agnostic-histology “baskets” seek the simultaneous detection of early efficacy signals among different histologies, to depict the influence that tumor lineage exerts in drug sensitivity. These studies can serve to interrogate uncharacterized genomic variants and to examine the effects of concurrent genomic alterations, whereas offering a real-time elucidation of acquired resistance mechanisms (67). Also, “basket” trials can unveil the toxicity effects of new drugs across multiple tumor types, allowing the investigators to have well-established safety profiles to support a favorable benefit:risk ratio in biomarker-selected patients. However, the obtained results must be interpreted with caution, as inferences from small number of patients might not be a secure source to make treatment-decisions.

Figure 2.

Integration of clinical-translational strategies into precision medicine drug development. Sophisticated master protocols are currently being used in the early drug development of new molecularly targeted drugs. “Umbrella” trials serve to study multiple targeted therapies in the context of a single disease (e.g., TRK/FGFR/RETinh in a specific tumor type). “Platform” trials have evolved as a more adaptable design, also aiming to study multiple targeted therapies in the context of a single disease but allowing therapies to enter or leave the platform on the basis of preclinical or clinical data becoming available throughout the study. “Basket” trials test a particular targeted therapy among patients that share a common molecular aberration (colored stars) across multiple cancer types (e.g., TRKinh in multiple histologies). These precision medicine approaches arise as the ideal scenario for implementing smart translational programs, based on the acquisition of biological samples from molecularly selected extraordinary responders. The collection of serial tumor biopsies and plasma samples, allowing the generation of patient-derived xenografts (PDX), has emerged as a powerful tool for integrating a deeper molecular knowledge from a specific target, fine-tuning optimal predictive biomarkers, early discovery of resistance mechanisms, and hypothesis generating of new therapeutic strategies. cfDNA, cell-free DNA.

Figure 2.

Integration of clinical-translational strategies into precision medicine drug development. Sophisticated master protocols are currently being used in the early drug development of new molecularly targeted drugs. “Umbrella” trials serve to study multiple targeted therapies in the context of a single disease (e.g., TRK/FGFR/RETinh in a specific tumor type). “Platform” trials have evolved as a more adaptable design, also aiming to study multiple targeted therapies in the context of a single disease but allowing therapies to enter or leave the platform on the basis of preclinical or clinical data becoming available throughout the study. “Basket” trials test a particular targeted therapy among patients that share a common molecular aberration (colored stars) across multiple cancer types (e.g., TRKinh in multiple histologies). These precision medicine approaches arise as the ideal scenario for implementing smart translational programs, based on the acquisition of biological samples from molecularly selected extraordinary responders. The collection of serial tumor biopsies and plasma samples, allowing the generation of patient-derived xenografts (PDX), has emerged as a powerful tool for integrating a deeper molecular knowledge from a specific target, fine-tuning optimal predictive biomarkers, early discovery of resistance mechanisms, and hypothesis generating of new therapeutic strategies. cfDNA, cell-free DNA.

Close modal

In this sense, regulatory agencies may have to consider changes in drug approval conditions, to deal with the inevitable degree of uncertainty associated with the efficacy evidence provided from trials lacking randomization, pooling across heterogeneous populations where sometimes there is no standard of care or limited data on prognosis. Accelerated approval translates that the magnitude of benefit-risk assessment performed by regulatory agencies is positive, but clinical evidence is incomplete. Feasible strategies to support these approvals should include temporal approvals or approvals at risk, that can be continuously reviewed once more data from post-authorization studies become available, reflecting the “real-world” data (68). These post-marketing data will be crucial for verifying the real clinical benefit of a new drug, specially when accelerated approvals are based on surrogate or intermediate endpoints, such as ORR instead of OS. Obviously, if benefit from new drugs is not verified when new information arises, indications should be withdrawn or modified accordingly. In parallel, collaborative efforts have focused on developing methods to establish the value of new specific cancer treatments, to support decision-making (69, 70).

Finally, new adaptive reimbursement strategies will need to be considered. Manufacturers could reduce the price of accelerated-approved drugs until robust efficacy confirmation is available. Confirmatory trials should be optimally designed for addressing the uncertainty of the real clinical effect of a new drug and conducted in a timely fashion, or confirmation could be provided from the data of expanded access programs (71).

Figure 3 summarizes some of the premises that should be carefully considered when pursuing the agnostic-histology development of new drugs.

Figure 3.

Considerations for agnostic-histology development of new drugs in oncology.

Figure 3.

Considerations for agnostic-histology development of new drugs in oncology.

Close modal

Moving away from traditional diagnostic criteria has translated into a new taxonomy of human diseases based on molecular biology. This has offered unique opportunities to more efficiently treat cancer patients, although it has brought several challenges. It seems clear that the positive experience with accelerated approvals in precision medicine will only continue if there is a collaborative effort between government, regulators, pharmaceutical and biotechnology industries, academia and patient groups (72).

We anticipate that agnostic-histology approvals will continue to grow and expand the therapeutic options for patients with cancer, because many other candidates are ready-to-go in the pipeline. We just need to be ready for embracing them and to calmly deal with the uncertainties while we try to minimize them.

C. Hierro reports receiving commercial research grants (to institution) from Bayer, speakers bureau honoraria from Ignyta and Lilly, and travel/accommodation grants from Lilly and Roche. J. Martin-Liberal reports receiving speakers bureau honoraria from Roche, Novartis, MSD, Pfizer, and Bristol-Myers Squibb, and travel grants from Ipsen. E. Garralda reports receiving commercial research grants from Novartis, speakers bureau honoraria from MSD, and is a consultant/advisory board member for Roche, Janssen, Boehringer, NeoMed Therapeutics, and Ellypses Pharma. No potential conflicts of interest were disclosed by the other authors.

The authors thank the support received from the Welfare Projects Division of the “La Caixa” Foundation to the Molecular Therapeutics Research Unit (UITM).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Hierro
C
,
Azaro
A
,
Argiles
G
,
Elez
E
,
Gomez
P
,
Carles
J
, et al
Unveiling changes in the landscape of patient populations in cancer early drug development
.
Oncotarget
2017
;
8
:
14158
72
.
2.
Martin-Liberal
J
,
Hierro
C
,
Ochoa de Olza
M
,
Rodon
J
. 
Immuno-oncology: the third paradigm in early drug development
.
Target Oncol
2017
;
12
:
125
38
.
3.
Hunter
DJ
. 
Uncertainty in the era of precision medicine
.
N Engl J Med
2016
;
375
:
711
3
.
4.
Kola
I
,
Bell
J
. 
A call to reform the taxonomy of human disease
.
Nat Rev Drug Discov
2011
;
10
:
641
2
.
5.
Mateo
J
,
Chakravarty
D
,
Dienstmann
R
,
Jezdic
S
,
Gonzalez-Perez
A
,
Lopez-Bigas
N
, et al
A framework to rank genomic alterations as targets for cancer precision medicine: the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT)
.
Ann Oncol
2018
;
29
:
1895
902
.
6.
Lacombe
D
,
Burock
S
,
Bogaerts
J
,
Schoeffski
P
,
Golfinopoulos
V
,
Stupp
R
. 
The dream and reality of histology agnostic cancer clinical trials
.
Mol Oncol
2014
;
8
:
1057
63
.
7.
Jhaveri
K
,
Juric
D
,
Saura
C
,
Cervantes
A
,
Melnyk
A
,
Patel
MR
, et al
A phase I basket study of the PI3K inhibitor taselisib (GDC-0032) in PIK3CA-mutated locally advanced or metastatic solid tumors [abstract]
. In:
Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14–18; Chicago, IL
.
Philadelphia (PA)
:
AACR
; 
2018
.
Cancer Res 2018;78(13 Suppl):CT046
.
8.
Flaherty
KT
,
Puzanov
I
,
Kim
KB
,
Ribas
A
,
McArthur
GA
,
Sosman
JA
, et al
Inhibition of mutated, activated BRAF in metastatic melanoma
.
N Engl J Med
2010
;
363
:
809
19
.
9.
Long
GV
,
Stroyakovskiy
D
,
Gogas
H
,
Levchenko
E
,
de Braud
F
,
Larkin
J
, et al
Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma
.
N Engl J Med
2014
;
371
:
1877
88
.
10.
Kopetz
S
,
Desai
J
,
Chan
E
,
Hecht
JR
,
O'Dwyer
PJ
,
Maru
D
, et al
Phase II pilot study of vemurafenib in patients with metastatic BRAF-mutated colorectal cancer
.
J Clin Oncol
2015
;
33
:
4032
8
.
11.
Prahallad
A
,
Sun
C
,
Huang
S
,
Di Nicolantonio
F
,
Salazar
R
,
Zecchin
D
, et al
Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR
.
Nature
2012
;
483
:
100
3
.
12.
Corcoran
RB
,
Ebi
H
,
Turke
AB
,
Coffee
EM
,
Nishino
M
,
Cogdill
AP
, et al
EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib
.
Cancer Discov
2012
;
2
:
227
35
.
13.
Corcoran
RB
,
Andre
T
,
Atreya
CE
,
Schellens
JHM
,
Yoshino
T
,
Bendell
JC
, et al
Combined BRAF, EGFR, and MEK Inhibition in Patients with BRAF(V600E)-Mutant Colorectal Cancer
.
Cancer Discov
2018
;
8
:
428
43
.
14.
Hyman
DM
,
Puzanov
I
,
Subbiah
V
,
Faris
JE
,
Chau
I
,
Blay
JY
, et al
Vemurafenib in multiple nonmelanoma cancers with BRAF V600 Mutations
.
N Engl J Med
2015
;
373
:
726
36
.
15.
Oneal
PA
,
Kwitkowski
V
,
Luo
L
,
Shen
YL
,
Subramaniam
S
,
Shord
S
, et al
FDA approval summary: vemurafenib for the treatment of patients with erdheim-chester disease with the BRAFV600 mutation
.
Oncologist
2018
;
23
:
1520
4
.
16.
Slamon
DJ
,
Leyland-Jones
B
,
Shak
S
,
Fuchs
H
,
Paton
V
,
Bajamonde
A
, et al
Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2
.
N Engl J Med
2001
;
344
:
783
92
.
17.
Bang
YJ
,
Van Cutsem
E
,
Feyereislova
A
,
Chung
HC
,
Shen
L
,
Sawaki
A
, et al
Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial
.
Lancet
2010
;
376
:
687
97
.
18.
Sartore-Bianchi
A
,
Trusolino
L
,
Martino
C
,
Bencardino
K
,
Lonardi
S
,
Bergamo
F
, et al
Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial
.
Lancet Oncol
2016
;
17
:
738
46
.
19.
Meric-Bernstam
F
,
Beeram
M
,
Mayordomo
JI
,
Hanna
DL
,
Ajani
JA
,
Blum
MA
, et al
Single agent activity of ZW25, a HER2-targeted bispecific antibody, in heavily pretreated HER2-expressing cancers
.
J Clin Oncol
36
, 
2018
(
suppl; abstr 2500
).
20.
Jhaveri
KL
,
Makker
V
,
Wang
XV
,
Chen
AP
,
Flaherty
K
,
Conley
BA
, et al
Ado-trastuzumab emtansine (T-DM1) in patients (pts) with HER2 amplified (amp) tumors excluding breast and gastric/gastro-esophageal junction (GEJ) adenocarcinomas: results from the National Cancer Institute (NCI) Molecular Analysis for Therapy Choice (MATCH) trial
.
J Clin Oncol
36
, 
2018
(
suppl; abstr 100
).
21.
Iwata
H
,
Tamura
K
,
Doi
T
,
Tsurutani
J
,
Modi
S
,
Park
H
, et al
Trastuzumab deruxtecan (DS-8201a) in subjects with HER2-expressing solid tumors: long-term results of a large phase 1 study with multiple expansion cohorts
.
J Clin Oncol
36
, 
2018
(
suppl; abstr 2501
).
22.
Cousin
S
,
Khalifa
E
,
Crombe
A
,
Laizet
Y
,
Lucchesi
C
,
Toulmonde
M
, et al
Targeting ERBB2 mutations in solid tumors: biological and clinical implications
.
J Hematol Oncol
2018
;
11
:
86
.
23.
Cabel
L
,
Fuerea
A
,
Lacroix
L
,
Baldini
C
,
Martin
P
,
Hollebecque
A
, et al
Efficacy of histology-agnostic and molecularly-driven HER2 inhibitors for refractory cancers
.
Oncotarget
2018
;
9
:
9741
50
.
24.
Hyman
DM
,
Piha-Paul
SA
,
Won
H
,
Rodon
J
,
Saura
C
,
Shapiro
GI
, et al
HER kinase inhibition in patients with HER2- and HER3-mutant cancers
.
Nature
2018
;
554
:
189
94
.
25.
Schumacher
TN
,
Schreiber
RD
. 
Neoantigens in cancer immunotherapy
.
Science
2015
;
348
:
69
74
.
26.
Snyder
A
,
Makarov
V
,
Merghoub
T
,
Yuan
J
,
Zaretsky
JM
,
Desrichard
A
, et al
Genetic basis for clinical response to CTLA-4 blockade in melanoma
.
N Engl J Med
2014
;
371
:
2189
99
.
27.
Rizvi
NA
,
Hellmann
MD
,
Snyder
A
,
Kvistborg
P
,
Makarov
V
,
Havel
JJ
, et al
Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer
.
Science
2015
;
348
:
124
8
.
28.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
29.
Le
DT
,
Durham
JN
,
Smith
KN
,
Wang
H
,
Bartlett
BR
,
Aulakh
LK
, et al
Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade
.
Science
2017
;
357
:
409
13
.
30.
Diaz
L
,
Marabelle
A
,
Kim
TW
,
Geva
R
,
Van Cutsem
E
,
André
T
, et al
Efficacy of pembrolizumab in phase 2 KEYNOTE-164 and KEYNOTE-158 studies of microsatellite instability high cancers
.
Ann Oncol
2017
;
28
(
suppl_5
).
Abstract nr 386P
.
31.
Muro
K
,
Chung
HC
,
Shankaran
V
,
Geva
R
,
Catenacci
D
,
Gupta
S
, et al
Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial
.
Lancet Oncol
2016
;
17
:
717
26
.
32.
Plimack
ER
,
Bellmunt
J
,
Gupta
S
,
Berger
R
,
Chow
LQ
,
Juco
J
, et al
Safety and activity of pembrolizumab in patients with locally advanced or metastatic urothelial cancer (KEYNOTE-012): a non-randomised, open-label, phase 1b study
.
Lancet Oncol
2017
;
18
:
212
20
.
33.
Nanda
R
,
Chow
LQ
,
Dees
EC
,
Berger
R
,
Gupta
S
,
Geva
R
, et al
Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 Study
.
J Clin Oncol
2016
;
34
:
2460
7
.
34.
Doi
T
,
Piha-Paul
SA
,
Jalal
SI
,
Saraf
S
,
Lunceford
J
,
Koshiji
M
, et al
Safety and antitumor activity of the anti-programmed death-1 antibody pembrolizumab in patients with advanced esophageal carcinoma
.
J Clin Oncol
2018
;
36
:
61
7
.
35.
O'Neil
BH
,
Wallmark
JM
,
Lorente
D
,
Elez
E
,
Raimbourg
J
,
Gomez-Roca
C
, et al
Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced colorectal carcinoma
.
PLoS One
2017
;
12
:
e0189848
.
36.
Rugo
HS
,
Delord
JP
,
Im
SA
,
Ott
PA
,
Piha-Paul
SA
,
Bedard
PL
, et al
Safety and antitumor activity of pembrolizumab in patients with estrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer
.
Clin Cancer Res
2018
;
24
:
2804
11
.
37.
Ott
PA
,
Bang
YJ
,
Berton-Rigaud
D
,
Elez
E
,
Pishvaian
MJ
,
Rugo
HS
, et al
Safety and antitumor activity of pembrolizumab in advanced programmed death ligand 1-positive endometrial cancer: results from the KEYNOTE-028 study
.
J Clin Oncol
2017
;
35
:
2535
41
.
38.
Bang
YJ
,
Doi
T
,
de Braud
F
,
Piha-Paul
S
,
Hollebecque
A
,
Abdul Razak
AR
, et al
525 Safety and efficacy of pembrolizumab (MK-3475) in patients (pts) with advanced biliary tract cancer: Interim results of KEYNOTE-028
.
Eur J Cancer
2015
;
51
Suppl 3
:
S112
.
39.
Pembrolizumab package insert
;
[cited 2018 Dec 23]. Available from
: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125514s014lbl.pdf.
40.
Hause
RJ
,
Pritchard
CC
,
Shendure
J
,
Salipante
SJ
. 
Classification and characterization of microsatellite instability across 18 cancer types
.
Nat Med
2016
;
22
:
1342
50
.
41.
Howitt
BE
,
Shukla
SA
,
Sholl
LM
,
Ritterhouse
LL
,
Watkins
JC
,
Rodig
S
, et al
Association of polymerase e-mutated and microsatellite-instable endometrial cancers with neoantigen load, number of tumor-infiltrating lymphocytes, and expression of PD-1 and PD-L1
.
JAMA Oncol
2015
;
1
:
1319
23
.
42.
Grasso
CS
,
Giannakis
M
,
Wells
DK
,
Hamada
T
,
Mu
XJ
,
Quist
M
, et al
Genetic mechanisms of immune evasion in colorectal cancer
.
Cancer Discov
2018
;
8
:
730
49
.
43.
Bouffet
E
,
Larouche
V
,
Campbell
BB
,
Merico
D
,
de Borja
R
,
Aronson
M
, et al
Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency
.
J Clin Oncol
2016
;
34
:
2206
11
.
44.
Geoerger
B
,
Jin Kang
H
,
Yalon-Oren
M
,
Marshall
LV
,
Vezina
C
,
Pappo
AS
, et al
KEYNOTE-051: an update on the phase 2 results of pembrolizumab (pembro) in pediatric patients (pts) with advanced melanoma or a PD-L1–positive advanced, relapsed or refractory solid tumor or lymphoma
.
J Clin Oncol
36
, 
2018
(
suppl; abstr 10525
).
45.
Kawakami
H
,
Zaanan
A
,
Sinicrope
FA
. 
Microsatellite instability testing and its role in the management of colorectal cancer
.
Curr Treat Options Oncol
2015
;
16
:
30
.
46.
Drilon
A
,
Laetsch
TW
,
Kummar
S
,
DuBois
SG
,
Lassen
UN
,
Demetri
GD
, et al
Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children
.
N Engl J Med
2018
;
378
:
731
9
.
47.
Larotrectinib package insert
;
[cited 2018 Dec 23]. Available from
: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/211710s000lbl.pdf.
48.
Lassen
UN
,
Albert
CM
,
Kummar
S
,
Van Tilburg
CM
,
Dubois
SG
,
Geoerger
B
, et al
Larotrectinib efficacy and safety in TRK fusion cancer: an expanded clinical dataset showing consistency in an age and tumor agnostic approach
.
Ann Oncol
2018
;
29
(
suppl_9
).
Abstract nr 4090
.
49.
Gao
Q
,
Liang
WW
,
Foltz
SM
,
Mutharasu
G
,
Jayasinghe
RG
,
Cao
S
, et al
Driver fusions and their implications in the development and treatment of human cancers
.
Cell Rep
2018
;
23
:
227
38
.e3.
50.
Demetri
GD
,
Paz-Ares
L
,
Farago
AF
,
Liu
SV
,
Chawla
SP
,
Tosi
D
, et al
Efficacy and safety of entrectinib in patients with NTRK fusion-positive (NTRK-fp) tumors: pooled analysis of STARTRK-2, STARTRK-1 and ALKA-372–001
.
Ann Oncol
2018
;
29
(
suppl_8
).
Abstract nr LBA17
.
51.
Drilon
A
,
Nagasubramanian
R
,
Blake
JF
,
Ku
N
,
Tuch
BB
,
Ebata
K
, et al
A next-generation TRK kinase inhibitor overcomes acquired resistance to prior TRK kinase inhibition in patients with TRK fusion-positive solid tumors
.
Cancer Discov
2017
;
7
:
963
72
.
52.
Drilon
A
,
Ou
SI
,
Cho
BC
,
Kim
DW
,
Lee
J
,
Lin
JJ
, et al
Repotrectinib (TPX-0005) is a next-generation ROS1/TRK/ALK inhibitor that potently inhibits ROS1/TRK/ALK solvent- front mutations
.
Cancer Discov
2018
;
8
:
1227
36
.
53.
Konicek
BW
,
Capen
AR
,
Credille
KM
,
Ebert
PJ
,
Falcon
BL
,
Heady
GL
, et al
Merestinib (LY2801653) inhibits neurotrophic receptor kinase (NTRK) and suppresses growth of NTRK fusion bearing tumors
.
Oncotarget
2018
;
9
:
13796
806
.
54.
Voss
MH
,
Hierro
C
,
Heist
RS
,
Cleary
JM
,
Meric-Bernstam
F
,
Gandhi
L
, et al
Debio 1347, an oral FGFR inhibitor: results from a first-in-human, phase I dose-escalation study in patients with FGFR genomically activated advanced solid tumors
.
J Clin Oncol
35
, 
2017
(
suppl; abstr 2500
).
55.
Bahleda
R
,
Meric-Bernstam
F
,
Goyal
L
,
Tran
B
,
He
H
,
Winkler
R
, et al
Phase I dose-escalation study of TAS-120, a highly selective, covalently bound FGFR inhibitor, in patients with advanced solid tumors [abstract]
. In:
Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14–18; Chicago, IL
.
Philadelphia (PA)
:
AACR
; 
2018
.
Cancer Res 2018;78(13 Suppl):CT121
.
56.
Drilon
AE
,
Subbiah
V
,
Oxnard
GR
,
Bauer
TM
,
Velcheti
V
,
Lakhani
NJ
, et al
A phase 1 study of LOXO-292, a potent and highly selective RET inhibitor, in patients with RET-altered cancers
.
J Clin Oncol
36
, 
2018
(
suppl; abstr 102
).
57.
Drilon
AE
,
Liu
S
,
Doebele
R
,
Rodriguez
C
,
Fakih
M
,
Reckamp
KL
, et al
A phase 1b study of RXDX-105, a VEGFR-sparing potent RET inhibitor, in RETi-naïve patients with RET fusion-positive NSCLC
.
Ann Oncol
2017
;
28
(
suppl_5
).
Abstract nr LBA19
.
58.
Subbiah
V
,
Gainor
JF
,
Rahal
R
,
Brubaker
JD
,
Kim
JL
,
Maynard
M
, et al
Precision targeted therapy with BLU-667 for RET-driven cancers
.
Cancer Discov
2018
;
8
:
836
49
.
59.
Garber
K
. 
Tissue-agnostic cancer drug pipeline grows, despite doubts
.
Nat Rev Drug Discov
2018
;
17
:
227
9
.
60.
Hellmann
MD
,
Ciuleanu
TE
,
Pluzanski
A
,
Lee
JS
,
Otterson
GA
,
Audigier-Valette
C
, et al
Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden
.
N Engl J Med
2018
;
378
:
2093
104
.
61.
Gandara
DR
,
Paul
SM
,
Kowanetz
M
,
Schleifman
E
,
Zou
W
,
Li
Y
, 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
;
24
:
1441
8
.
62.
Ciardiello
F
,
Arnold
D
,
Casali
PG
,
Cervantes
A
,
Douillard
JY
,
Eggermont
A
, et al
Delivering precision medicine in oncology today and in future-the promise and challenges of personalised cancer medicine: a position paper by the European Society for Medical Oncology (ESMO)
.
Ann Oncol
2014
;
25
:
1673
8
.
63.
Woodcock
J
,
LaVange
LM
. 
Master protocols to study multiple therapies, multiple diseases, or both
.
N Engl J Med
2017
;
377
:
62
70
.
64.
Mullard
A
. 
NCI-MATCH trial pushes cancer umbrella trial paradigm
.
Nat Rev Drug Discov
2015
;
14
:
513
5
.
65.
Calvo
F
,
Apolone
G
,
Baumann
M
,
Caldas
C
,
Celis
JE
,
de Lorenzo
F
, et al
Cancer Core Europe: a European cancer research alliance realizing a research infrastructure with critical mass and programmatic approach to cure cancer in the 21st century
.
Eur J Cancer
2018
;
103
:
155
9
.
66.
Hanker
AB
,
Brewer
MR
,
Sheehan
JH
,
Koch
JP
,
Sliwoski
GR
,
Nagy
R
, et al
An acquired HER2(T798I) gatekeeper mutation induces resistance to neratinib in a patient with HER2 mutant-driven breast cancer
.
Cancer Discov
2017
;
7
:
575
85
.
67.
Tao
JJ
,
Schram
AM
,
Hyman
DM
. 
Basket studies: redefining clinical trials in the era of genome-driven oncology
.
Annu Rev Med
2018
;
69
:
319
31
.
68.
Gellad
WF
,
Kesselheim
AS
. 
Accelerated approval and expensive drugs - a challenging combination
.
N Engl J Med
2017
;
376
:
2001
4
.
69.
Schnipper
LE
,
Davidson
NE
,
Wollins
DS
,
Tyne
C
,
Blayney
DW
,
Blum
D
, et al
American Society of Clinical Oncology statement: a conceptual framework to assess the value of cancer treatment options
.
J Clin Oncol
2015
;
33
:
2563
77
.
70.
Cherny
NI
,
Sullivan
R
,
Dafni
U
,
Kerst
JM
,
Sobrero
A
,
Zielinski
C
, et al
A standardised, generic, validated approach to stratify the magnitude of clinical benefit that can be anticipated from anti-cancer therapies: the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS)
.
Ann Oncol
2015
;
26
:
1547
73
.
71.
Martin Algarra
S
,
Soriano
V
,
Fernandez-Morales
L
,
Berciano-Guerrero
MA
,
Mujika
K
,
Manzano
JL
, et al
Dabrafenib plus trametinib for compassionate use in metastatic melanoma: a STROBE-compliant retrospective observational postauthorization study
.
Medicine
2017
;
96
:
e9523
.
72.
Lemery
S
,
Keegan
P
,
Pazdur
R
. 
First FDA approval agnostic of cancer site—when a biomarker defines the indication
.
N Engl J Med
2017
;
377
:
1409
12
.