3,207
Views
5
CrossRef citations to date
0
Altmetric
Oncology

Transforming approaches to treating TRK fusion cancer: historical comparison of larotrectinib and histology-specific therapies

, , , , &
Pages 59-70 | Received 16 Sep 2020, Accepted 03 Nov 2020, Published online: 28 Nov 2020

Abstract

Objective

The results from basket trials utilized to gain regulatory approval of tumor-agnostic therapies can be difficult to interpret without the context of a comparator arm. We describe the role and efficacy of histology-based treatments to provide a historical comparison with larotrectinib.

Methods

A systematic literature review (SLR) was conducted on the clinical outcomes of current histology-based standard of care treatments used in non-small cell lung cancer, colorectal cancer, thyroid cancer, gliomas, soft tissue sarcoma, salivary gland cancer, and infantile fibrosarcoma (7 of the 21 tumor histologies in the larotrectinib trials). The review focused on advanced stage/metastatic disease to make a historical comparison with larotrectinib.

Results

Larotrectinib provides positive outcomes in both adult and pediatric patients with advanced or metastatic solid tumors known to harbor NTRK gene fusions across a wide range of tumor types. Although the numbers of patients per tumor type are limited, the results of this historical comparison demonstrated that larotrectinib is an efficacious treatment option when naïvely indirectly compared with historical treatments across all 7 reviewed tumor types, especially in comparison to later lines of therapy.

Conclusions

Utilizing larotrectinib as a case example across these types of historical comparisons shows that larotrectinib provides positive efficacy outcomes in TRK fusion cancer across tumor histologies known to harbor NTRK gene fusions that may be preferable to historical treatments.

Introduction

The tropomyosin receptor kinase (TRK) inhibitor, larotrectinib, marks the first European Medicines Agency (EMA) approval of a tumor-agnostic therapyCitation1. Larotrectinib is a potent and specific inhibitor of all three TRK proteins: TRKA, TRKB, and TRKCCitation2. In addition to the EMA, the Food and Drug Administration (FDA), Agência Nacional de Vigilância Sanitária (ANVISA), Health Canada (HC), Taiwan Food and Drug Administration (TFDA), Saudi Food and Drug Administration (SFDA), and the Swiss Agency for Therapeutic Products (Swissmedic) have approved larotrectinib for use in adult or pediatric patients with solid tumors that display a neurotrophic tyrosine receptor kinase (NTRK) gene fusion who have disease that is locally advanced, metastatic, or where surgical resection is likely to result in severe morbidity, and who have no satisfactory treatment optionsCitation3. Larotrectinib is the first drug to be approved with a tumor-agnostic indication as the first and only indication. Since the approval of larotrectinib, entrectinib has also received approval in the US and Japan for the treatment of adult and pediatric patients (≥12 years of age) with solid tumors that have an NTRK gene fusion without a known acquired resistance mutation, are metastatic or where surgical resection is likely to result in severe morbidity, and have progressed following treatment or have no satisfactory alternative therapyCitation4.

Although the frequency of NTRK gene fusions varies by tumor localization, NTRK gene fusions occur in only a few thousand patients in the European Union (EU) annually, meeting the criteria of an ultra-rare diseaseCitation5. Rare primary genomic alterations, such as NTRK gene fusions, pose unique problems to clinical research programs. The randomized controlled trial (RCT) design is considered methodologically the gold standard; however, in cases of a rare oncogenic driver where the prevalence in any single tumor histology is extremely low, this study design would face significant enrollment challengesCitation6,Citation7. Besides the low number of patients available for recruitment, the choice of comparator arm challenges the feasibility of designing an RCT, particularly when the targetable alteration is spread across a wide range of tumor types, all of which differ in natural history and treatment optionsCitation7. For these reasons, there is a need to use novel, adaptive study designs to advance the drug development processCitation6,Citation8,Citation9. One such design is the single-arm basket trial; in this trial design, patients who have the same molecular feature, regardless of their cancer histology, are enrolled. If efficacy and safety are identified across tumor histologies in early trials, the basket trial design becomes the registrational trial design. The EMA has recognized the need for and validity of clinical trials with adaptive characteristics in the clinical development of novel therapiesCitation10.

However, clinical interpretation of the results from basket trials can be challenging due to the histological heterogeneity of the patient populationCitation11. Further, because of the single arm design, there cannot be a defined standard of care treatment compared across various tumor histologies; therefore, making time-to-event endpoints, such as survival, difficult to interpret and extrapolate to the clinical settingCitation12. Despite these challenges, basket trials are being used in the drug regulatory review and approval process, and there needs to be a way to incorporate such tissue agnostic therapies into clinical practiceCitation11. As such, the question becomes, how does an oncology clinician take the data from an adaptive trial and apply them within a single histology to make them applicable to the patient populations they are treating? Herein, we use the example of the agnostic (histology-independent) development of larotrectinib, the first targeted therapy for NTRK gene fusions, to describe for the oncology practitioner one method of evaluating outcomes from the larotrectinib basket trial relative to historical, histology-based treatments. Such historical comparisons against the novel treatment approach help to put the relative clinical outcomes of larotrectinib in context for the provider. This review seeks to summarize the efficacy of treatments for select TRK fusion tumor histologies in order to conduct a historical comparison with larotrectinib.

Methods

Literature review of historical treatments for TRK fusion cancer

Historically, NTRK gene fusions in patients with solid tumor cancer were not routinely screened for or identified. In the minority of patients with a known NTRK gene fusion at diagnosis, treatment was initiated per guideline recommendations for the specific tumor histology and disease stage, where available, rather than with a therapy targeted to the oncogenic driver because of the lack of targeted treatment options available for these patients prior to the availability of larotrectinib or entrectinib. TRK fusion cancer was historically treated with a selection of chemotherapy or, potentially, biologic therapy or immunotherapy, based largely upon tumor histologyCitation13–15.

In order to provide a thorough assessment of the historical treatments for TRK fusion cancer, a systematic literature review (SLR) evaluating patient populations within each of the relevant tumor histologies and stage of disease appropriate for larotrectinib treatment was conducted. The goal of the SLR was to provide historical data on which a generalized naïve indirect comparison of larotrectinib against current treatment approaches or standard of care could be based. The SLR excluded any historical molecularly targeted therapies due to the mutual exclusivity of oncogenic driversCitation16. The SLR was conducted for 21 histologies and in accordance with the United Kingdom (UK) National Institute for Health and Care and Excellence (NICE) guidance and Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidance. Bibliographic databases including MEDLINE (via PubMed), Embase (via Embase.com), and the Cochrane Library were searched for relevant studies published from database inception to February/March 2019. The database searches used medical subheading (MeSH) and Emtree index terms as well as free text terms appropriate for each database and included relevant terms to identify each patient population of interest based on tumor histology. Additional terms for specific comparators of interest, clinical outcomes, or study designs were included for some tumor histologies with a larger body of available published evidence to focus the SLR on the most pertinent data. The full search strategies for each tumor type are provided in the Supplementary Appendix. Grey literature sources, including conference abstracts and clinical trial registries, also were hand-searched for relevant information. Study selection criteria describing the population, interventions/comparators, outcomes, and study designs of interest for each SLR were defined in an SLR protocol developed a priori. The complete SLR protocols are available upon request from the authors. Dual screening of the literature and quality assessment of all included studies were performed by two researchers, and all data extraction was independently validated.

Tumor type and stage selection

The tumor histologies reported here represent a subset of the 21 histologies included in the larotrectinib clinical development program and include non-small cell lung cancer (NSCLC), colorectal cancer (CRC), thyroid cancer, gliomas, soft tissue sarcoma (STS) (excluding gastrointestinal stromal tumor [GIST]), salivary gland cancer, and infantile fibrosarcoma (IFS). These seven tumor types were chosen as representing both the more frequently occurring tumor types with a low reported frequency of NTRK gene fusions (i.e. NSCLC, CRC) as well as those more rare tumors which have a higher reported frequency of NTRK gene fusions (i.e. thyroid cancer, salivary gland cancer, and IFS) (). Additionally, non-GIST STS and gliomas were included as representative of a high unmet needs patient population, as these tumor types largely lack therapies that target specific oncogenic drivers. The historical data drawn from the SLR are used here to highlight the importance of this type of data in approved therapies utilizing adaptive clinical trial designs. The focus is on an “all-comer” (defined as all patients within a specific histology) patient population because NTRK gene fusions were not routinely screened for or identified historically prior to the approval of larotrectinib. The review also focused on advanced or metastatic disease, as this stage of disease is more representative of the TRK fusion cancer population included in the larotrectinib clinical development program. Additionally, the historical treatments included for comparison were based on tumor type and also aligned to the line of therapy in which larotrectinib would be used based on its labeled indication, which states that larotrectinib is indicated for use in pediatric and adult patients with solid tumors that harbor an NTRK gene fusion and are metastatic or where surgical resection is likely to result in severe morbidity, and have no satisfactory or alternative treatments, or that have progressed following treatmentCitation3; in practice, this generally means later lines of therapy for those tumor types with established standard of care treatment options.

Table 1. Global incidence and NTRK gene fusion frequency amongst the 7 tumor types included in this analysis.

Results

Clinical outcomes of historical treatment for specific tumor histologies that harbor NTRK gene fusions

In prevalent solid tumor cancer histologies harboring NTRK gene fusions, such as NSCLC and CRC, the histology-based treatment paradigm in the metastatic setting is continually evolving, but it is generally well-established and guideline-based. The efficacy of historical treatments for NSCLC and CRC is shown in . Despite improved outcomes with first-line treatment of NSCLC, the overall response rate (ORR) for patients with second-line or subsequent therapy utilizing histology-based treatment in metastatic NSCLC is <30%. Further, the median progression-free survival (PFS) in this same population is <6 months. Although outcomes are similar for metastatic CRC, with a median PFS ranging from 1.4 months to 13.2 months for second-line or subsequent therapy, ORR worsens through each line of therapy. In patients receiving third-line or subsequent therapy, the ORR reported ranges from 1% to 13% compared with 11% to 47.7% in the second-line setting ().

Table 2. Efficacy of historical tumor histology-specific treatments.

Solid tumor cancer histologies that harbor NTRK gene fusions and occur with a lower prevalence, including radioiodine (RAI)-refractory thyroid, glioma, STS (non-GIST), and salivary gland, are generally treated with biological therapy (non-molecularly targeted therapies such as vascular endothelial growth factor [VEGF] inhibitors), molecularly targeted therapy, or chemotherapy. The efficacy of historical treatments for thyroid and salivary gland cancer are shown in . In metastatic RAI-refractory thyroid cancer, the histology-based therapies include chemotherapy and VEGF inhibition. Although VEGF inhibitors are frequently used in this setting, the ORR in the first-line or subsequent setting is poor, ranging from 0% to 64.8% (). This same treatment pattern is commonly employed for gliomas, with VEGF inhibitors being used as single agent or in combination with chemotherapy. Although these glioma therapies can produce higher ORR (range: 63–95.2%), the median PFS in this population is poor, with the majority of studies reporting <6 months in the second or subsequent line of therapy. In patients without targetable oncogenic drivers with STS (non-GIST), the therapies for metastatic disease primarily consist of chemotherapy. Even in the first-line setting, the ORR for chemotherapy ranges from 17.2% to 44.4%, with the ORR decreasing to 13.2% as additional lines of therapy are included. Salivary gland cancer is not well studied, as the majority of data come from small, prospective studies. However, in these studies, the ORR in metastatic salivary gland cancer for chemotherapy is 31% in the first-line setting and decreases to 5% in the second-line setting. Further, the median PFS for the first-line setting is only 6 months ().

Rare solid tumor cancer histologies that harbor NTRK gene fusions, including IFS, differ from the above-mentioned histologies as metastatic spread of disease is uncommon, and most patients are considered curative with surgical resection alone, although 3 of 50 infants have considerable morbidity associated with surgery, including amputationCitation139. Due to this risk for significant morbidity associated with surgery, many IFS patients require neoadjuvant treatmentCitation139. The ORR associated with neoadjuvant chemotherapy + surgery ranges from 71% to 88.9%, leaving room for improvement in this pediatric population ().

Tumor histology-based clinical outcomes comparison of historical treatment vs larotrectinib

Clinical trials for larotrectinib enrolled pediatric and adult patients with TRK fusion cancer across solid tumor histologies. The rarity of TRK fusion cancer, in addition to the lack of equipoise in tumor histologies without available standard therapies or where recommended therapies exist but fail to provide a documented and relevantly sized clinical benefit, and the expectations for patient cross-over (if an RCT were conducted) made it not feasible or appropriate to conduct a randomized trial to demonstrate improvement in overall survival (OS). Therefore, the results from the larotrectinib trials were pooled to provide evidence of efficacy and safety per regulatory request. The data presented here are drawn from two separate data cutoffs of the pooled analysis data for larotrectinib. The first dataset, including 93 patients with solid tumor and nine patients with primary central nervous system (CNS) tumors, is independent review committee-assessed data with a data cutoff date of 30 July 2018 that were used for the EMA submission and approval; the ORR for the total population (N = 102, including patients with solid tumors and primary CNS tumors) was 67%Citation3. The second dataset, which included the 93 patients with solid tumors from the 30 July 2018 data cutoff plus an additional 66 patients with TRK fusion solid tumors, is investigator-assessed data published by Hong et al in 2020 with a data cutoff date of 19 February 2019Citation31. The ORR for the total population (N = 159, including only patients with solid tumors [no primary CNS tumors included in this analysis]) was 79%Citation31. Each of these datasets has response rate data available by primary tumor type as outlined in .

When comparing within specific tumor histologies, larotrectinib remains an efficacious treatment option compared with historical treatments. Within NSCLC, the ORR observed for the 12 patients with NSCLC who received larotrectinib was 75%; this is higher than what has been reported to date within the historical treatment setting, as ORR in the second-line or beyond treatment setting has been reported up to 29% (). This same trend was reported in both salivary gland tumors (n = 20) and non-GIST STS (n = 36), where the ORR was 90% and 81%, respectively. Historically, the ORR for salivary gland tumors has only been reported up to 31%, and this was in the first-line setting, with second-line therapy reporting an ORR of only 5% (). In non-GIST STS, the historical ORR across lines of therapy has ranged from 13.2% to 44.4% in the first-line setting or beyond (). In CRC, there are more well-established treatment options across multiple lines of therapy; however, the response rates in the third-line setting or beyond range from 0% to 13%, falling short of the 50% response rate reported for CRC patients with NTRK gene fusions treated with larotrectinib (n = 8) ().

Discussion

Clinical trials for larotrectinib enrolled pediatric and adult patients with TRK fusion cancer across solid tumor histologiesCitation2,Citation30,Citation140. The rarity of TRK fusion cancer, in addition to the lack of equipoise in tumor histologies without available standard therapies or where recommended therapies exist but fail to provide a documented and relevantly sized clinical benefit, and the expectations for patient cross-over (if an RCT were conducted), made it not feasible or appropriate to conduct an RCT to demonstrate improvement in OSCitation141. Furthermore, given the large number of primary tumor types that have different natural histories, it was not scientifically appropriate to “lump” these tumor types together into a single randomized trialCitation141. Therefore, the results from the larotrectinib trials were pooled to provide evidence of efficacy and safety per regulatory requestCitation141.

Utilizing larotrectinib as the case example across these types of historical comparisons shows that larotrectinib provides positive efficacy outcomes in TRK fusion cancer across tumor histologies known to harbor NTRK gene fusions that may be preferable to historical treatments. As the case example, larotrectinib has shown consistent efficacy and safety in TRK fusion cancer across multiple tumor types in both pediatric and adult patientsCitation2,Citation30,Citation140. Historical therapies for tumor histologies that harbor NTRK gene fusions, although lacking data specifically in TRK fusion cancer, have highly variable efficacy (). Utilizing historical data provides for a non-statistical side-by-side comparison of data that is histology-specific; comparing the data in this manner shows that larotrectinib appears to be more efficacious within these tumor types compared with historical treatments.

Safety was not a part of this review, but is an important consideration when making therapeutic decisions. Many patients with TRK fusion cancer were historically treated with chemotherapy per guideline recommendations, the adverse event (AE) profiles of which can be detrimental to patient’s quality of life, leading to both short-term and long-term toxicitiesCitation142,Citation143. A patient’s ability or willingness to tolerate such AEs, rather than uncontrolled disease or lack of potential active anticancer therapy, may rapidly become the limiting factor for treatment successCitation143. TRK inhibitors offer patients an efficacious targeted therapy option with a favorable tolerability profileCitation3,Citation144,Citation145.

As precision medicine in oncology continues to progress, an increasing number of studies with adaptive designs, such as basket trials, will be conductedCitation9. There are multiple tumor-agnostic therapies in development, including merestinib, TPX-0005, and selitrectinib (LOXO-195), that are being studied in an adaptive design clinical development program. Larotrectinib is one of the first agents to be studied in a tumor-agnostic manner and receive approval in the United States (US)—and the first to receive approval in the EU; as such, it provides clinicians the first opportunity to understand the clinical development program and basis for comparison for these agentsCitation3,Citation143. Since the approval of larotrectinib, entrectinib has also received approval based on data from a basket trial in the USCitation4. Although both larotrectinib and entrectinib are approved in a tumor-agnostic manner for TRK fusion cancer, larotrectinib is a highly selective TRK inhibitor, whereas entrectinib is a multi-kinase inhibitor with lower specificity for TRK and also specificity for ROS1 and ALKCitation3,Citation4,Citation144.

There are some drawbacks associated with this type of historical comparison. First, only ORR data is available for larotrectinib by tumor type. Due to both the small number of patients in each tumor type, and the study design for larotrectinib (single-arm, basket trial), time-to-event endpoints such as PFS and OS are not adequately characterized by tumor type. Secondly, this is just a general comparison of the outcomes of therapies in unselected, tumor histology-based patient populations vs the outcomes reported for larotrectinib across tumor histologies. These outcomes were not matched based on any demographic or clinical parameters. Third, the numbers of patients within each tumor histology included in the larotrectinib clinical trials are low, making meaningful comparisons to historical outcomes more difficult. Also, within the comparison of response rate data, it should be noted that in the first published pooled analysis of larotrectinib including 55 patients with TRK fusion cancer, acquired drug resistance was noted in 10 patients; acquired drug resistance was defined as disease progression during treatment after a documented objective response or stable diseaseCitation2. However, the impact of acquired drug resistance on treatment outcomes remains unknown. Finally, as stated previously, we do not know if any of the patients enrolled in the standard of care treatments had NTRK gene fusion-positive tumors; this is important as information pertaining to the prognosis of this patient population in relation to other genotypes or wild-type tumors is currently not available. There is limited evidence to suggest that NTRK gene fusions are predictive of poorer outcomesCitation146–149; however, there are currently no long-term studies specifically following TRK fusion-positive cancer patients. A recent retrospective review identified 76 cases of TRK fusion-positive cancer across 17 distinct tumor types; the ORR across all first-line therapies (exclusive of TRK inhibitor therapy) in this population was 46.7% (n = 7/15)Citation150. Further, the ORR was 62.5% (n = 15/24) for those patients who received chemotherapy across all lines of therapy for advanced disease and 11.1% (n = 1/12) for those who received immunotherapy across all lines of therapy for advanced diseaseCitation150. It is difficult to garner outcomes from this review as the population sizes are small for efficacy data; however, patients with TRK fusion-positive cancers may respond to alternative standards of care, although efficacy of immunotherapy in the absence of other predictive biomarkers (i.e. microsatellite instability-high) appears limitedCitation150.

When translating basket trial data into clinical practice, historical treatment comparisons are just one way in which to understand the efficacy of a tumor-agnostic therapy across tumor histology types. Other options exist, including intrapatient comparison and tools/scales developed to assess the benefit of therapies. A retrospective exploratory intra-patient comparison analysis using successive time to progression (TTP) as a way to detect whether a new agent is having a modulating effect on tumor growth effectively uses a patient as their own control. If a new agent has an anti-tumor effect, it will change the natural history of the disease; so, if TTPn is greater than TTPn-1, then it is likely that the new agent is having an effect on the natural history of that patient’s tumor. Growth modulation index (GMI) is the ratio of the TTPn and TTPn-1, and GMI ≥ 1.33 was defined as a sign of clinical activity by Von HoffCitation151. Further, the European Society for Medical Oncology (ESMO) developed a validated and reproducible tool to assess the magnitude of the clinical benefit for cancer therapiesCitation152. Per the Magnitude of Clinical Benefit Scale (MCBS), single-arm trials in orphan disease states or those with a high unmet need that show an ORR > 60%, or a median PFS > 6 months, or an ORR ≥ 20% to <60% and a duration of response ≥9 months are considered to have the highest magnitude of clinical benefit based on a preliminary scoreCitation153. This preliminary score is further adjusted based on toxicity (downgrade 1 level if there are ≥30% grade 3–4 toxicities affecting daily well-being), quality of life (QoL) (upgrade 1 level if improved QoL), or confirmatory phase 4 experience (upgrade 1 level for confirmatory, adequately sized, phase 4 experience)Citation153. Use of an intrapatient analysis or validated tools allows both clinicians and decision makers to gain a greater understanding of the place in therapy of a drug for which there is no comparator arm in clinical trials.

Conclusions

In a tumor-agnostic scenario, many individual and rare tumor histologies are brought together with a common oncogenic driver. As more novel therapeutics are being studied in a tumor-agnostic manner, there will be an increasing need for level-setting the data produced from these studies with data that have been reported for historically available therapies across tumor histologies. There is no perfect method for understanding the clinical impact of these therapies in specific tumor histologies, and it is likely many different methodologies for comparison will emerge as we move further into the era of precision medicine. A few methodologies likely to play key roles in the interpretation of these data across tumor histologies include historical comparisons and intra-patient comparison of TTP/PFS on successive lines of therapyCitation151,Citation152,Citation154. Although historical comparisons are general comparisons that do not provide statistical comparative efficacy, these types of comparisons do provide clinicians with side-by-side data for understanding the place in therapy of a novel tumor-agnostic therapy within a specific tumor histology. However, it should be noted that the status of genomic alteration is usually not known in historical studies.

In this review, we highlighted the historical treatment data for specific tumor histologies and compared them to the data from the larotrectinib basket trial. Although the numbers of patients are small, the ORR of larotrectinib is higher than what has been historically reported across most specific tumor histologies in the line of therapy where larotrectinib is most likely to be used based on the approved label; the approval for larotrectinib is in patients with TRK fusion cancer with locally advanced or metastatic disease or where surgical resection is likely to result in severe morbidity and who have no satisfactory treatment optionsCitation3. These data provide clinicians with an understanding of where larotrectinib may best fit into treatment paradigms in specific tumor histologies. Based on its efficacy from the basket trial, larotrectinib is now included as a recommended therapy across a range of solid tumor histologies, as denoted by inclusion in 19 National Comprehensive Cancer Network (NCCN) guidelines. Considering the growing complexities of precision medicine along with increasing variation in clinical trial design, both clinicians and healthcare decision makers will need to assess therapies to determine their clinical benefit vs currently used therapies. It becomes imperative that we strive to create a reasonable basis for comparisons for these therapies so that patients not only gain access to them but also that clinicians and healthcare decision makers have an understanding of where these therapies would provide the greatest benefit in the treatment course.

Transparency

Declaration of funding

Bayer Healthcare Pharmaceuticals, Inc.

Declaration of financial/other relationships

M Pollack is an employee of Xcenda, which received sponsorship from Bayer for the research and manuscript development. K Keating is an employee of Bayer HealthCare Pharmaceuticals Inc. E Wissinger is an employee of Xcenda, which received sponsorship from Bayer for the research and manuscript development. L Jackson is an employee of Bayer HealthCare Pharmaceuticals Inc. E Sarnes is an employee of Xcenda, which received sponsorship from Bayer for the research and manuscript development. B Cuffel is an employee of Bayer HealthCare Pharmaceuticals Inc. Peer reviewers on this manuscript have received an honorarium from CMRO for their review work but have no other relevant financial relationships to disclose.

Authors’ contributions/authorship

MP helped design the SLR and subsequent targeted review and helped write the manuscript. KK helped design the SLR and helped write the manuscript. EW designed the SLR and helped write the manuscript. ES helped design the SLR and helped write the manuscript. BC helped design the SLR and helped write the manuscript.

Data deposition

NA.

Supplemental material

Supplemental Material

Download MS Word (43.6 KB)

Acknowledgements

The authors would like to acknowledge the help of Kylie Matthews in the medical writing process.

Data availability statement

Review article; no data/results provided in this publication.

References

  • European Medicines Agency (EMA). Vitrakvi (larotrectinib) [Internet]. [cited 2019 Oct 11]. Available from: https://www.ema.europa.eu/en/medicines/human/summaries-opinion/vitrakvi.
  • Drilon A, Laetsch TW, Kummar S, et al. Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med. 2018;378(8):731–739.
  • VITRAKVI® (larotrectinib) [summary of product characteristics (SmPC)]. Whippany (NJ): Bayer HealthCare Pharmaceuticals, Inc.; 2019.
  • Meldolesi J. Neurotrophin TRK receptors: new targets for cancer therapy. Rev Physiol Biochem Pharmacol. 2018;174:67–79.
  • Food and Drug Administration (FDA). FDA approves entrectinib for NTRK solid tumors and ROS-1 NSCLC [Internet]. [cited 2019 Oct 10]. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-entrectinib-ntrk-solid-tumors-and-ros-1-nsclc.
  • Panageas KS. Clinical trial design for rare cancers: why a less conventional route may be required. Expert Rev Clin Pharmacol. 2015;8(6):661–663.
  • Bogaerts J, Sydes MR, Keat N, et al. Clinical trial designs for rare diseases: studies developed and discussed by the International Rare Cancers Initiative. Eur J Cancer. 2015;51(3):271–281.
  • Schram AM, Hyman DM. Quantifying the benefits of genome-driven oncology. Cancer Discov. 2017;7(6):552–554.
  • Jarvis LM. Cancer, redefined. Chem Eng News. 2015;95:26–30.
  • European Medicines Agency (EMA). Reflection paper on methodological issues in confirmatory clinical trials planned with an adaptive design [Internet]. 2007. [cited 2020 Jan 23]. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/reflection-paper-methodological-issues-confirmatory-clinical-trials-planned-adaptive-design_en.pdf.
  • Dubuque C, Major P, Suarez S, et al. Oncology basket trials: an emerging paradigm shift in trial design & treatment approach? Trinity Partners; 2018. https://trinitylifesciences.com/wp-content/uploads/2019/05/Oncology_Basket_Trials_-_Trinity_Partners.pdf
  • United States Food & Drug Administration. Clinical trial endpoints for the approval of cancer drugs and biologics. 2018. [cited 2020 Oct 9]. Available from: https://www.fda.gov/media/71195/download.
  • Novello S, Barlesi F, Califano R, et al. Metastatic non-small-cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016;27(suppl 5):v1–v27.
  • ESMO NSCLC. eUpdate 28 June 2017: Treatment Recommendations and Revised ESMO Magnitude of Clinical Benefit Scale (ESMO-MCBS) grading for metastatic non-small-cell lung cancer and eUpdate 28 June 2017: New eUpdate featuring updated treatment algorithms for metastatic non-small-cell lung cancer [Internet]. [cited 2018 Jul 17]. Available from: https://www.esmo.org/Guidelines/Lung-and-Chest-Tumours/Metastatic-Non-Small-Cell-Lung-Cancer.
  • Van Cutsem E, Cervantes A, Adam R, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27(8):1386–1422.
  • Jiao X, Lokker A, Snider J, et al. Co-occurrence of NTRK fusions with other genomic biomarkers in cancer patients. European Society for Medical Oncology Congress, 2019, Sept 27–Oct 1, Barcelona, Spain.
  • Globocan. Cancer fact sheets. 2018. [cited 2020 Oct 9]. http://gco.iarc.fr/today/fact-sheets-cancers?cancer=7&type=0&sex=0.
  • Farago AF, Taylor MS, Doebele RC, et al. Clinicopathologic features of non-small-cell lung cancer harboring an NTRK gene fusion. J Clin Oncol Precis Oncol. 2018;2018(2):1–12.
  • Stransky N, Cerami E, Schalm S, et al. The landscape of kinase fusions in cancer. Nat Commun. 2014;5:4846.
  • Sassolas G, Hafdi-Nejjari Z, Ferraro A, et al. Oncogenic alterations in papillary thyroid cancers of young patients. Thyroid. 2012;22(1):17–26.
  • Bongarzone I, Vigneri P, Mariani L, et al. RET/NTRK1 rearrangements in thyroid gland tumors of the papillary carcinoma family: correlation with clinicopathological features. Clin Cancer Res. 1998;4(1):223–228.
  • Leeman-Neill RJ, Kelly L, Liu P, et al. ETV6-NTRK3 is a common chromosomal rearrangement in radiation-associated thyroid cancer. Cancer. 2014;120(6):799–807.
  • Brzeziańska E, Karbownik M, Migdalska-Sek M, et al. Molecular analysis of the RET and NTRK1 gene rearrangements in papillary thyroid carcinoma in the Polish population. Mutat Res. 2006;599(1–2):26–35.
  • Prasad ML, Vyas M, Horne MJ, et al. NTRK fusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States. Cancer. 2016;122(7):1097–1107.
  • Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol. 2018;15(12):731–747.
  • Skalova A, Vanecek T, Martinek P, et al. Molecular profiling of mammary analog secretory carcinoma revealed a subset of tumors harboring a novel ETV6-RET translocation: report of 10 cases. Am J Surg Pathol. 2018;42(2):234–246.
  • Ferrari A, Orbach D, Sultan I, et al. Neonatal soft tissue sarcomas. Semin Fetal Neonatal Med. 2012;17(4):231–238.
  • Bourgeois JM, Knezevich SR, Mathers JA, et al. Molecular detection of the ETV6-NTRK3 gene fusion differentiates congenital fibrosarcoma from other childhood spindle cell tumors. Am J Surg Pathol. 2000;24(7):937–946.
  • Pavlick D, Schrock AB, Malicki D, et al. Identification of NTRK fusions in pediatric mesenchymal tumors. Pediatr Blood Cancer. 2017;64(8):e26433.
  • Davis JL, Lockwood CM, Albert CM, et al. Infantile NTRK-associated mesenchymal tumors. Pediatr Dev Pathol. 2018;21(1):68–78.
  • Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol. 2020;21(4):531–540.
  • Drilon A, DuBois SG, Farago AF, et al. Activity of larotrectinib in TRK fusion cancer patients with brain metastases or primary central nervous system tumors. Presented at American Society of Clinical Oncology Annual Meeting, May 31–June 4, 2019, Chicago, IL.
  • Doz F, Geoerger B, DuBois S, et al. RARE-45. Activity of larotrectinib in TRK fusion cancer patients with primary central nervous system tumors. Neuro-Oncology. 2019;21(Supplement_6):vi231–vi231.
  • Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373(2):123–135.
  • Lena H, Rizvi NA, Wolf J, et al. Nivolumab in patients (pts) with advanced refractory squamous (SQ) non-small cell lung cancer (NSCLC): 2-year follow-up from CheckMate 063 and exploratory cytokine profiling analyses. J Thorac Oncol. 2016;11(4):S115–S116.
  • Spigel D, Reckamp K, Rizvi N, et al. A phase III study (CheckMate 017) of nivolumab (NIVO; antiprogrammed death-1) vs docetaxel (DOC) in previously treated advanced or metastatic squamous (SQ) cell non-small cell lung cancer (NSCLC). J Clin Oncol. 2015;33(15_suppl):8009–8009.
  • Borghaei H, Brahmer JR, Horn L, et al. Nivolumab (nivo) vs docetaxel (doc) in patients (pts) with advanced NSCLC: CheckMate 017/057 2-y update and exploratory cytokine profile analyses. J Clin Oncol. 2016;34(15_suppl):9025–9025.
  • Font EF, Gettinger SN, Burgio MA, et al. Three-year follow-up from CheckMate 017/057: nivolumab versus docetaxel in patients with previously treated advanced non-small cell lung cancer (NSCLC). Ann Oncol. 2017;28:v462.
  • Horn L, Spigel DR, Vokes EE, et al. Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung cancer: two-year outcomes from two randomized, open-label, phase III trials (CheckMate 017 and CheckMate 057). J Clin Oncol. 2017;35(35):3924–3933.
  • Thomas M, Kohlhaufl M, Reck M, et al. Long-term outcomes with nivolumab in patients with previously treated advanced non-small cell lung cancer (NSCLC). Oncol Res Treat. 2017;40:137.
  • Garon E, Ciuleanu T, Arrieta O, et al. Ramucirumab plus docetaxel versus placebo plus docetaxel for second-line treatment of stage IV non-small-cell lung cancer after disease progression on platinum-based therapy (REVEL): a multicentre, double-blind, randomised phase 3 trial. Lancet. 2014;384(9944):665–673.
  • Bidoli PCF, Favaretto A, Alabiso O, et al. Update of REVEL: a randomized, double-blind, phase III study of docetaxel (DOC) and ramucirumab (RAM; IMC-1121B) versus DOC and placebo (PL) in the second-line (2L) treatment of stage IV non-small cell lung cancer (NSCLC) including subgroup analysis of histology. Ann Oncol. 2015;26(Suppl 6):vi73–vi89.
  • Gervais R, Ducolone A, Breton JL, et al. Phase II randomised trial comparing docetaxel given every 3 weeks with weekly schedule as second-line therapy in patients with advanced non-small-cell lung cancer (NSCLC). Ann Oncol. 2005;16(1):90–96.
  • Gridelli C, Gallo C, Maio MD, et al. A randomised clinical trial of two docetaxel regimens (weekly vs 3 week) in the second-line treatment of non-small-cell lung cancer. The DISTAL 01 study. Br J Cancer. 2004;91(12):1996–2004.
  • Hanna N, Shepherd FA, Fossella FV, et al. Randomized phase III trial of pemetrexed versus docetaxel in patients with non-small-cell lung cancer previously treated with chemotherapy. J Clin Oncol. 2004;22:1589–1597.
  • Demarinis F, Paul S, Hanna N, et al. Survival update for the phase III study of pemetrexed vs docetaxel in non-small cell lung cancer (NSCLC). J Clin Oncol. 2006;24(18_suppl):7133–7133.
  • Lai CL, Tsai CM, Chiu CH, et al. Phase II randomized trial of tri-weekly versus days 1 and 8 weekly docetaxel as a second-line treatment of advanced non-small cell lung cancer. Jpn J Clin Oncol. 2005;35(12):700–706.
  • Manegold C, Pilz LR, Koschel G, et al. Randomized multicenter phase II study of gemcitabine versus docetaxel as first-line therapy with second-line crossover in advanced-stage non-small-cell lung cancer. Clin Lung Cancer. 2005;7(3):208–214.
  • Quoix E, Lebeau B, Depierre A, et al. Randomised, multicentre phase II study assessing two doses of docetaxel (75 or 100 mg/m2) as second-line monotherapy for non-small-cell lung cancer. Ann Oncol. 2004;15(1):38–44.
  • Schuette W, Nagel S, Blankenburg T, et al. Phase III study of second-line chemotherapy for advanced non-small-cell lung cancer with weekly compared with 3-weekly docetaxel. J Clin Oncol. 2005;23(33):8389–8395.
  • Sun Y, Wu YL, Zhou CC, et al. Second-line pemetrexed versus docetaxel in Chinese patients with locally advanced or metastatic non-small cell lung cancer: a randomized, open-label study. Lung Cancer. 2013;79(2):143–150.
  • Vergnenegre A, Corre R, Berard H, et al. Cost-effectiveness of second-line chemotherapy for non-small cell lung cancer: an economic, randomized, prospective, multicenter phase III trial comparing docetaxel and pemetrexed: the GFPC 05-06 study. J Thorac Oncol. 2011;6(1):161–168.
  • Hosomi Y, Yoh K, Kasahara K, et al. Docetaxel + ramucirumab (DR) versus docetaxel + placebo (D) as second-line treatment for advanced non-small cell lung cancer (NSCLC): a randomized, phase II, double-blind, multicenter trial in Japan. J Clin Oncol. 2015;33(15_suppl):8054–8054.
  • Yoh K, Hosomi Y, Kasahara K, et al. A randomized, double-blind, phase II study of ramucirumab plus docetaxel vs placebo plus docetaxel in Japanese patients with stage IV non-small cell lung cancer after disease progression on platinum-based therapy. Lung Cancer. 2016;99:186–193.
  • Bradbury PA, Morris DG, Nicholas G, et al. Canadian Cancer Trials Group (CCTG) IND211: a randomized trial of pelareorep (Reolysin) in patients with previously treated advanced or metastatic non-small cell lung cancer receiving standard salvage therapy. Lung Cancer. 2018;120:142–148.
  • Li R, Sun L, Wang J, et al. Pemetrexed versus docetaxel in second line non-small-cell lung cancer: results and subsets analyses of a multi-center, randomized, exploratory trial in Chinese patients. Pulm Pharmacol Ther. 2012;25(5):364–370.
  • Manegold C, Koschel G, Hruska D, et al. Open, randomized, phase II study of single-agent gemcitabine and docetaxel as first- and second-line treatment in patients with advanced non-small-cell lung cancer. Clin Lung Cancer. 2007;8(4):245–251.
  • Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–1639.
  • Camps C, Massuti B, Jiménez A, et al. Randomized phase III study of 3-weekly versus weekly docetaxel in pretreated advanced non-small-cell lung cancer: a Spanish lung cancer group trial. Ann Oncol. 2006;17(3):467–472.
  • Cullen MH, Zatloukal P, Sörenson S, et al. A randomized phase III trial comparing standard and high-dose pemetrexed as second-line treatment in patients with locally advanced or metastatic non-small-cell lung cancer. Ann Oncol. 2008;19(5):939–945.
  • Fehrenbacher L, Spira A, Ballinger M, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet. 2016;387(10030):1837–1846.
  • Mazières J, Park K, Lewanski C, et al. 3-Year survival and duration of response in randomized phase II study of atezolizumab (atezo) vs docetaxel (doc) in 2L + NSCLC (POPLAR. J Thorac Oncol. 2018;13(4):S79.
  • Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540–1550.
  • Herbst RS, Baas P, Kim DW, et al. Pembrolizumab (pembro) vs docetaxel (Doce) for previously treated, PD-L1-expressing NSCLC: updated outcomes of KEYNOTE-010. Ann Oncol. 2016;27:1–36.
  • Ohe Y, Ichinose Y, Nakagawa K, et al. Efficacy and safety of two doses of pemetrexed supplemented with folic acid and vitamin B12 in previously treated patients with non-small cell lung cancer. Clin Cancer Res. 2008;14(13):4206–4212.
  • Cortinovis D, Pawel JV, Syrigos K, et al. Immune-related adverse events (irAEs) in advanced NSCLC patients treated with atezolizumab: safety population analyses from the Ph III study OAK. Ann Oncol. 2017;28:v468.
  • Fehrenbacher L, Pawel JV, Park K, et al. Updated efficacy analysis including secondary population results for OAK: a randomized phase III study of atezolizumab versus docetaxel in patients with previously treated advanced non-small cell lung cancer. J Thorac Oncol. 2018;13(8):1156–1170.
  • Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389(10066):255–265.
  • Satouchi M, Fehrenbacher L, Cobo DM, et al. Long-term survival in atezolizumab-treated patients with 2l + NSCLC from Ph III randomized OAK study. J Thorac Oncol. 2017;12(11):S1794.
  • Shepherd FA, Dancey J, Ramlau R, et al. Prospective randomized trial of docetaxel versus best supportive care in patients with non-small-cell lung cancer previously treated with platinum-based chemotherapy. J Clin Oncol. 2000;18(10):2095–2103.
  • Dong L, Han ZF, Feng ZH, et al. Comparison of pemetrexed and docetaxel as salvage chemotherapy for the treatment for nonsmall-cell lung cancer after the failure of epidermal growth factor receptor-tyrosine kinase inhibitors. J Int Med Res. 2014;42(1):191–197.
  • Chen Y-M, Shih J-F, Perng R-P, et al. A randomized trial of different docetaxel schedules in non-small-cell lung cancer patients who failed previous platinum-based chemotherapy. Chest. 2006;129(4):1031–1038.
  • Spigel DR, McLeod M, Hussein MA, et al. Randomized results of fixed-duration (1-yr) vs continuous nivolumab in patients (pts) with advanced non-small cell lung cancer (NSCLC). Ann Oncol. 2017;28:v461.
  • Ducreux M, Malka D, Mendiboure J, et al. Sequential versus combination chemotherapy for the treatment of advanced colorectal cancer (FFCD 2000-05): an open-label, randomised, phase 3 trial. Lancet Oncol. 2011;12(11):1032–1044.
  • Van Cutsem E, Tabernero J, Lakomy R, et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol. 2012;30(28):3499–3506.
  • Joulain F, Proskorovsky I, Allegra C, et al. Mean overall survival gain with aflibercept plus FOLFIRI vs placebo plus FOLFIRI in patients with previously treated metastatic colorectal cancer. Br J Cancer. 2013;109(7):1735–1743.
  • Clarke SJ, Yip S, Brown C, et al. Single-agent irinotecan or FOLFIRI as second-line chemotherapy for advanced colorectal cancer; results of a randomised phase II study (DaVINCI) and meta-analysis [corrected]. Eur J Cancer. 2011;47(12):1826–1836.
  • Tabernero J, Yoshino T, Cohn AL, et al. Ramucirumab versus placebo in combination with second-line FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first-line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): a randomised, double-blind, multicentre, phase 3 study. Lancet Oncol. 2015;16(5):499–508.
  • Masi G, Salvatore L, Boni L, et al. Continuation or reintroduction of bevacizumab beyond progression to first-line therapy in metastatic colorectal cancer: final results of the randomized BEBYP trial. Ann Oncol. 2015;26(4):724–730.
  • Cao R, Zhang S, Ma D, et al. A multi-center randomized phase II clinical study of bevacizumab plus irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI) compared with FOLFIRI alone as second-line treatment for Chinese patients with metastatic colorectal cancer. Med Oncol. 2015;32(1):325.
  • Galal KM, Abdelsalam M, Fawzy EE, et al. Role of bevacizumab as post-progression maintenance therapy in metastatic colon cancer. J Med Sci. 2008;8(5):452–460.
  • Fuchs CS, Moore MR, Harker G, et al. Phase III comparison of two irinotecan dosing regimens in second-line therapy of metastatic colorectal cancer. J Clin Oncol. 2003;21(5):807–814.
  • Iwamoto S, Takahashi T, Tamagawa H, et al. FOLFIRI plus bevacizumab as second-line therapy in patients with metastatic colorectal cancer after first-line bevacizumab plus oxaliplatin-based therapy: the randomized phase III EAGLE study. Ann Oncol. 2015;26(7):1427–1433.
  • Giantonio BJ, Catalano PJ, Meropol NJ, et al. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200. J Clin Oncol. 2007;25(12):1539–1544.
  • Cunningham D, Glimelius B. A phase III study of irinotecan (CPT-11) versus best supportive care in patients with metastatic colorectal cancer who have failed 5-fluorouracil therapy. V301 Study Group. Semin Oncol. 1999;26:6–12.
  • Cunningham D, Pyrhonen S, James RD, et al. Randomised trial of irinotecan plus supportive care versus supportive care alone after fluorouracil failure for patients with metastatic colorectal cancer. Lancet. 1998;352(9138):1413–1418.
  • Kim GP, Sargent DJ, Mahoney MR, et al. Phase III noninferiority trial comparing irinotecan with oxaliplatin, fluorouracil, and leucovorin in patients with advanced colorectal carcinoma previously treated with fluorouracil: N9841. J Clin Oncol. 2009;27(17):2848–2854.
  • Rougier P, Lepille D, Bennouna J, et al. Antitumour activity of three second-line treatment combinations in patients with metastatic colorectal cancer after optimal 5-FU regimen failure: a randomised, multicentre phase II study. Ann Oncol. 2002;13(10):1558–1567.
  • Rothenberg ML, Oza AM, Bigelow RH, et al. Superiority of oxaliplatin and fluorouracil-leucovorin compared with either therapy alone in patients with progressive colorectal cancer after irinotecan and fluorouracil-leucovorin: interim results of a phase III trial. J Clin Oncol. 2003;21(11):2059–2069.
  • Grothey A, Van Cutsem E, Sobrero A, et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 2013;381(9863):303–312.
  • Ricotta R, Verrioli A, Ghezzi S, et al. Radiological imaging markers predicting clinical outcome in patients with metastatic colorectal carcinoma treated with regorafenib: post hoc analysis of the CORRECT phase III trial (RadioCORRECT study). ESMO Open. 2017;1(6):e000111.
  • Xu J, Kim TW, Shen L, et al. Results of a randomized, double-blind, placebo-controlled, phase III trial of trifluridine/tipiracil (TAS-102) monotherapy in Asian patients with previously treated metastatic colorectal cancer: the TERRA study. J Clin Oncol. 2018;36(4):350–358.
  • Kim TW, Shen L, Xu JM, et al. TERRA: a randomized, double-blind, placebo-controlled phase 3 study of TAS-102 in Asian patients with metastatic colorectal cancer. Ann Oncol. 2016;27:465PD.
  • Yoshino T, Mizunuma N, Yamazaki K, et al. TAS-102 monotherapy for pretreated metastatic colorectal cancer: a double-blind, randomised, placebo-controlled phase 2 trial. Lancet Oncol. 2012;13(10):993–1001.
  • Yoshino T, Shinozaki E, Yamazaki K, et al. Final survival results and onset of neutropenia as an indicator of therapeutic effect in phase 2 of TAS-102 vs placebo with metastatic colorectal cancer (J003-10040030). Ann Oncol. 2016;27:ii107.
  • Kemeny N, Garay CA, Gurtler J, et al. Randomized multicenter phase II trial of bolus plus infusional fluorouracil/leucovorin compared with fluorouracil/leucovorin plus oxaliplatin as third-line treatment of patients with advanced colorectal cancer. J Clin Oncol. 2004;22(23):4753–4761.
  • Mayer RJ, Van Cutsem E, Falcone A, et al. Randomized trial of TAS-102 for refractory metastatic colorectal cancer. N Engl J Med. 2015;372(20):1909–1919.
  • Mayer RJ, Ohtsu A, Yoshino T, et al. TAS-102 versus placebo plus best supportive care in patients with metastatic colorectal cancer refractory to standard therapies: final survival results of the phase III RECOURSE trial. J Clin Oncol. 2016;34(4_suppl):634–634.
  • Li J, Qin S, Xu R, et al. Regorafenib plus best supportive care versus placebo plus best supportive care in Asian patients with previously treated metastatic colorectal cancer (CONCUR): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2015;16(6):619–629.
  • Brose MS, Nutting CM, Jarzab B, et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet. 2014;384(9940):319–328.
  • Dadu R, Waguespack SG, Sherman SI, et al. Efficacy and tolerability of different starting doses of sorafenib in patients with differentiated thyroid cancer. Oncologist. 2014;19(5):477–482.
  • Onoda N, Sugino K, Higashiyama T, et al. The safety and efficacy of weekly paclitaxel administration for anaplastic thyroid cancer patients: a nationwide prospective study. Thyroid. 2016;26(9):1293–1299.
  • Ain KB, Egorin MJ, De Simone PA. Treatment of anaplastic thyroid carcinoma with paclitaxel: phase 2 trial using ninety-six-hour infusion. Collaborative Anaplastic Thyroid Cancer Health Intervention Trials (CATCHIT) Group. Thyroid. 2000;10(7):587–594.
  • Higashiyama T, Ito Y, Hirokawa M, et al. Induction chemotherapy with weekly paclitaxel administration for anaplastic thyroid carcinoma. Thyroid. 2010;20(1):7–14.
  • Akl FMF, Eladawei GE, Abd-Alkhalek SE, et al. Do concurrent chemoradiotherapy with docetaxel followed by docetaxel consolidation chemotherapy improve the outcome of anaplastic thyroid carcinoma patients? Forum Clin Oncol. 2013;4:19–24.
  • Ito Y, Onoda N, Ito KI, et al. Sorafenib in Japanese patients with locally advanced or metastatic medullary thyroid carcinoma and anaplastic thyroid carcinoma. Thyroid. 2017;27(9):1142–1148.
  • Takahashi S, Kiyota N, Yamazaki T, et al. A phase II study of the safety and efficacy of lenvatinib in patients with advanced thyroid cancer. Future Oncol. 2019;15(7):717–726.
  • Takahashi S, Kiyota N, Yamazaki T, et al. Phase II study of lenvatinib in patients with differentiated, medullary, and anaplastic thyroid cancer: final analysis results. J Clin Oncol. 2016;34(15_suppl):6088–6088.
  • Bible KC, Suman VJ, Menefee ME, et al. A multiinstitutional phase 2 trial of pazopanib monotherapy in advanced anaplastic thyroid cancer. J Clin Endocrinol Metab. 2012;97(9):3179–3184.
  • Schlumberger M, Tahara M, Wirth LJ, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N Engl J Med. 2015;372(7):621–630.
  • Catalano MG, Pugliese M, Gallo M, et al. Valproic acid, a histone deacetylase inhibitor, in combination with paclitaxel for anaplastic thyroid cancer: results of a multicenter randomized controlled phase II/III trial. Int J Endocrinol. 2016;2016:2930414.
  • Sosa JA, Elisei R, Jarzab B, et al. Randomized safety and efficacy study of fosbretabulin with paclitaxel/carboplatin against anaplastic thyroid carcinoma. Thyroid. 2014;24(2):232–240.
  • Kim JH, Leeper RD. Treatment of locally advanced thyroid carcinoma with combination doxorubicin and radiation therapy. Cancer. 1987;60(10):2372–2375.
  • Savvides P, Nagaiah G, Lavertu P, et al. Phase II trial of sorafenib in patients with advanced anaplastic carcinoma of the thyroid. Thyroid. 2013;23(5):600–604.
  • Mooney CJ, Nagaiah G, Fu P, et al. A phase II trial of fosbretabulin in advanced anaplastic thyroid carcinoma and correlation of baseline serum-soluble intracellular adhesion molecule-1 with outcome. Thyroid. 2009;19(3):233–240.
  • Schaub C, Tichy J, Schafer N, et al. Prognostic factors in recurrent glioblastoma patients treated with bevacizumab. J Neurooncol. 2016;129(1):93–100.
  • Vredenburgh JJ, Desjardins A, Herndon JE, et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007;13(4):1253–1259.
  • McDonald CR, Delfanti RL, Krishnan AP, et al. Restriction spectrum imaging predicts response to bevacizumab in patients with high-grade glioma. Neuro Oncol. 2016;18(11):1579–1590.
  • Raval S, Hwang S, Dorsett L. Bevacizumab and irinotecan in patients (pts) with recurrent glioblastoma multiforme (GBM). J Clin Oncol. 2007;25(18_suppl):2078–2078.
  • Omuro A, Chan TA, Abrey LE, et al. Phase II trial of continuous low-dose temozolomide for patients with recurrent malignant glioma. Neuro Oncol. 2013;15(2):242–250.
  • Hassenbusch SJ, Nardone EM, Levin VA, et al. Stereotactic injection of DTI-015 into recurrent malignant gliomas: phase I/II trial. Neoplasia. 2003;5(1):9–16.
  • Gkogkou P, Geropantas K, Ajithkumar T. An individualized-approach to second-line systemic anti-cancer therapy for glioblastoma. Ann Oncol. 2017;28:v117.
  • Shahid T, Bhattacharyya GS, Malhotra H, et al. Continuous low dose temozolamide with or without VT-122 in glioblastoma. J Clin Oncol. 2015;33(15_suppl):e13010–e13010.
  • Brada M, Judson I, Beale P, et al. Phase I dose-escalation and pharmacokinetic study of temozolomide (SCH 52365) for refractory or relapsing malignancies. Br J Cancer. 1999;81(6):1022–1030.
  • Estlin EJ, Lashford L, Ablett S, et al. Phase I study of temozolomide in paediatric patients with advanced cancer. United Kingdom Children's Cancer Study Group. Br J Cancer. 1998;78(5):652–661.
  • Hundsberger T, Brugge D, Putora PM, et al. Re-irradiation with and without bevacizumab as salvage therapy for recurrent or progressive high-grade gliomas. J Neurooncol. 2013;112(1):133–139.
  • Mong S, Ellingson BM, Nghiemphu PL, et al. Persistent diffusion-restricted lesions in bevacizumab-treated malignant gliomas are associated with improved survival compared with matched controls. AJNR Am J Neuroradiol. 2012;33(9):1763–1770.
  • Kreisl TN, Zhang W, Odia Y, et al. A phase II trial of single-agent bevacizumab in patients with recurrent anaplastic glioma. Neuro Oncol. 2011;13(10):1143–1150.
  • Sardi I, Cetica V, Massimino M, et al. Promoter methylation and expression analysis of MGMT in advanced pediatric brain tumors. Oncol Rep. 2009;22(4):773–779.
  • Airoldi M, Garzaro M, Pedani F, et al. Cisplatin + vinorelbine treatment of recurrent or metastatic salivary gland malignancies (RMSGM): a final report on 60 cases. Am J Clin Oncol. 2017;40(1):86–90.
  • Widemann BC, Reinke DK, Helman LJ, et al. SARC006: phase II trial of chemotherapy in sporadic and neurofibromatosis type 1 (NF1)-associated high-grade malignant peripheral nerve sheath tumors (MPNSTs). J Clin Oncol. 2013;31(15_suppl):10522–10522.
  • Higham CS, Steinberg SM, Dombi E, et al. SARC006: phase II trial of chemotherapy in sporadic and neurofibromatosis type 1 associated chemotherapy-naive malignant peripheral nerve sheath tumors. Sarcoma. 2017;2017:8685638.
  • Kroep JR, Ouali M, Gelderblom H, et al. First-line chemotherapy for malignant peripheral nerve sheath tumor (MPNST) versus other histological soft tissue sarcoma subtypes and as a prognostic factor for MPNST: an EORTC soft tissue and bone sarcoma group study. Ann Oncol. 2011;22(1):207–214.
  • Weiss B, Widemann BC, Wolters P, et al. Sirolimus for progressive neurofibromatosis type 1-associated plexiform neurofibromas: a neurofibromatosis Clinical Trials Consortium phase II study. Neuro Oncol. 2015;17(4):596–603.
  • Lamm W, Schur S, Kostler WJ, et al. Clinical signs of neurofibromatosis impact on the outcome of malignant peripheral nerve sheath tumors. Oncology. 2014;86(2):122–126.
  • Yoo KH, Kim HS, Lee SJ, et al. Efficacy of pazopanib monotherapy in patients who had been heavily pretreated for metastatic soft tissue sarcoma: a retrospective case series. BMC Cancer. 2015;15:154.
  • Orbach D, Rey A, Cecchetto G, et al. Infantile fibrosarcoma: management based on the European experience. J Clin Oncol. 2010;28(2):318–323.
  • Kynaston JA, Malcolm AJ, Craft AW, et al. Chemotherapy in the management of infantile fibrosarcoma. Med Pediatr Oncol. 1993;21(7):488–493.
  • Orbach D, Brennan B, De Paoli A, et al. Conservative strategy in infantile fibrosarcoma is possible: the European Paediatric Soft Tissue Sarcoma Study Group experience. Eur J Cancer. 2016;57:1–9.
  • Laetsch TW, DuBois SG, Mascarenhas L, et al. Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol. 2018;19(5):705–714.
  • Food and Drug Administration (FDA). NDA multidisciplinary review and evaluation NDA 210861 and NDA 211710: VITRAKVI (larotrectinib) [Internet]. [cited 2019 Oct 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/210861Orig1s000_211710Orig1s000MultidisciplineR.pdf.
  • Lopategui DM, Yechieli R, Ramasamy R. Oncofertility in sarcoma patients. Transl Androl Urol. 2017;6(5):951–958.
  • Palumbo MO, Kavan P, Miller WH, Jr, et al. Systemic cancer therapy: achievements and challenges that lie ahead. Front Pharmacol. 2013;4:57.
  • Vitrakvi [prescribing information]. Whippany (NJ): Bayer Healthcare Pharmaceuticals Inc.; 2018.
  • Rozlytrek [prescribing information]. South San Francisco (CA): Genentech USA, Inc.; 2019.
  • Musholt TJ, Musholt PB, Khaladj N, et al. Prognostic significance of RET and NTRK1 rearrangements in sporadic papillary thyroid carcinoma. Surgery. 2000;128(6):984–993.
  • Lassaletta A, Zapotocky M, Bouffet E, et al. An integrative molecular and genomic analysis of pediatric hemispheric low-grade gliomas: an update. Childs Nerv Syst. 2016;32(10):1789–1797.
  • Gooskens SL, Houwing ME, Vujanic GM, et al. Congenital mesoblastic nephroma 50 years after its recognition: a narrative review. Pediatr Blood Cancer. 2017;64(7):e26437.
  • Pietrantonio F, Di Nicolantonio F, Schrock AB, et al. ALK, ROS1, and NTRK rearrangements in metastatic colorectal cancer. J Natl Cancer Inst. 2017;109:12.
  • Rosen EY, Goldman DA, Hechtman JF, et al. TRK fusions are enriched in cancers with uncommon histologies and the absence of canonical driver mutations. Clin Cancer Res. 2020;26(7):1624–1632.
  • Von Hoff DD. There are no bad anticancer agents, only bad clinical trial designs––21st Richard and Hinda Rosenthal Foundation Award Lecture. Clin Cancer Res. 1998;4(5):1079–1086.
  • Cherny NI, Sullivan R, Dafni U, 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(8):1547–1573.
  • Cherny NI, Dafni U, Bogaerts J, et al. ESMO-Magnitude of Clinical Benefit Scale version 1.1. Ann Oncol. 2017;28(10):2340–2366.
  • Massard C, Michiels S, Ferte C, et al. High-throughput genomics and clinical outcome in hard-to-treat advanced cancers: results of the MOSCATO 01 trial. Cancer Discov. 2017;7(6):586–595.