Larotrectinib

Evaluating larotrectinib for the treatment of advanced solid tumors harboring an NTRK gene fusion

Roberto Filippi, Ilaria Depetris & Maria Antonietta Satolli

To cite this article: Roberto Filippi, Ilaria Depetris & Maria Antonietta Satolli (2021) Evaluating larotrectinib for the treatment of advanced solid tumors harboring an NTRK gene fusion, Expert Opinion on Pharmacotherapy, 22:6, 677-684, DOI: 10.1080/14656566.2021.1876664
To link to this article: https://doi.org/10.1080/14656566.2021.1876664

Published online: 12 Feb 2021. Submit your article to this journal

Article views: 141
View related articles View Crossmark data

Full Terms & Conditions of access and use can be found at https://www.tandfonline.com/action/journalInformation?journalCode=ieop20

EXPERT OPINION ON PHARMACOTHERAPY 2021, VOL. 22, NO. 6, 677–684

https://doi.org/10.1080/14656566.2021.1876664

DRUG EVALUATION
Evaluating larotrectinib for the treatment of advanced solid tumors harboring an NTRK gene fusion
Roberto Filippi a, Ilaria Depetris b and Maria Antonietta Satollic
aMedical Oncology 1 – AOU Città Della Salute E Della Scienza Di Torino; Candiolo Cancer Institute, FPO – IRCCS Candiolo; Department of Oncology, University of Turin, Turin, Italy; bMedical Oncology, Ospedale Civile Di Ivrea, Ivrea, Turin, Italy; cMedical Oncology 1 – AOU Città Della Salute E Della Scienza Di Torino; Department of Oncology, University of Turin, Turin, Italy

ARTICLE HISTORY
Received 7 October 2020
Accepted 12 January 2021
KEYWORDS
NTRK; trk; larotrectinib; loxo- 101; gene fusions; rare cancers; precision oncology

1.Introduction
Despite harboring multiple genetic, epigenetic, and chromo- somal abnormalities, oncogene-addicted cancer cells show a marked dependency on one or few genes for survival and growth. Tumors harboring mutated oncogenic drivers are dependent on the aberrant activity of the driver, and thus show sensitivity to the targeting of the driver, which is con- sidered a convenient development strategy of new antineo- plastic agents. In this setting, in fact, specific targeted inhibitors can overcome the intricate and heterogeneous remaining mutational landscape of the tumor. Indeed, this approach led to the approval of successful drugs against the protein products of genes such as cKIT, EGFR, HER2, and BRAF.

2.The NTRK pathway
The three Neurotrophic Tropomyosin Receptor Kinase (NTRK) proto-oncogenes (NTRK1, NTRK2, NTRK3) encode the three members of the Trk receptor family (TrkA, TrkB, TrkC, respec- tively). Primarily involved in the physiology of neuronal devel- opment and homeostasis, these genes are expressed at high concentration in neuronal cells and neural crest-derived cells. However, some isoforms are expressed in non-neural tissues as well [1,2]. The binding with their cognate ligands, repre- sented by various neurotrophins [3,4], entails the homodimer- ization and transactivation of the receptor kinase. The downstream intracellular signaling cascade ultimately leads

to neuronal proliferation, differentiation and survival [5], as well as to synapse strengthening [4]. The involved pathways vary with the specific Trk receptor and isoform, and include Ras/MAPK, phospholipase C-gamma, and PI3K/Akt [4,6] (Figure 1).
The genetic alteration that links NTRK genes to tumori- genesis is mainly represented by the genetic fusion, described with tens of different partners [7,8]. The resulting fusion product provides an aberrant downstream mitogenic and antiapoptotic signal. NTRK fusions are rare events in carcinogenesis, that occur in 1% of cancer cases [9], with marked heterogeneity across primary sites. Their frequency is highest among some rare adult tumors (incidence 75% to over 90%), such as secretory breast cancer, mammary-analog secretory carcinoma of the salivary gland, as well as in relatively common pediatric and neonatal tumors, including infantile fibrosarcoma, and cellular congenital mesoblastic nephroma. These adults malignancies are characterized solely by NTRK3 fusions, whereas pediatric cancers also by NTRK1 rearrangements [10–12]. A lower incidence (5 to 25%) is observed among other infrequent malignancies, as papil- lary thyroid cancer, Spitz tumor, the so-called ‘wild-type’ gastrointestinal stromal tumor, and pediatric high-grade gliomas. In addition, NTRK fusions are present at low fre- quency (<5%, often <1%) in a wide range of common adult and pediatric malignancies [13,14] (Table 1). Among other

CONTACT Roberto Filippi [email protected] Medical Oncology 1 - AOU Città Della Salute E Della Scienza Di Torino; Candiolo Cancer Institute, FPO - IRCCS Candiolo; Department of Oncology, University of Turin, Corso Bramante 88, Turin, 10126, Italy.
© 2021 Informa UK Limited, trading as Taylor & Francis Group

oncogenic NTRK alterations, mutations [15] and at least one splice variant (TrkAIII) [16] are also described. TrkAIII exhibits skipping of exons 6, 7, and 9, resulting in deletion of a specific receptor extracellular domain, which physiologi- cally prevents spontaneous activation of the protein. This oncogenic mechanism could be allowed or enhanced by NTRK1 gene amplification or overexpression [16].

3.Overview of the market
Cancer is a widespread, growing disease, coming with healthcare and social costs. Even limited niches of patients

Figure 1. Trk signaling pathways and cellular effects of its activation. In italics the effects of physiology-only interest. BDGF, brain-derived growth factor; DAG, diacylglycerol; IP3, inositol trisphosphate; NT, neurotrophin; NGF, nerve growth factor.

Box 1. Drug summary box.
Drug name Larotrectinib

Phase Launched
Indication NTRK fusion-positive cancers
Pharmacological Description Potent and selective inhibitor of NTRK fusion protein products
Route of Administration Oral
Chemical structure

Pivotal trial NAVIGATE phase-2 trial (NCT02576431)

Table 1. Main primary sites that may feature NTRK fusions.

High-grade glioma Pancreatic cancer Head and neck cancer Colorectal cancer
Lung cancer Renal cell carcinoma
Breast cancer Gastrointestinal stromal tumor Cholangiocarcinoma Pediatric and adult sarcomas
Melanoma, Spitzoid tumors Pediatric inflammatory myofibroblastic tumor Thyroid cancer Pediatric low-grade gliomas
Salivary gland cancer Congenital mesoblastic nephroma

potentially constitute large populations in absolute num- bers. Oncogene addiction frequently limits prognosis and response to traditional agents [17–19], and an unfavorable prognostic impact is specifically suggested for NTRK fusions [20,21]. Hence, the need of patients with cancers harboring gene fusions to be spared toxicities from sub- optimal treatments [19] and to receive newer, effective drugs [22–25].
These drugs mostly block the mitogenic signaling cascade at the receptor level. Trk is part of the target spectrum of several multikinase inhibitors, including crizotinib, nintedanib and pona- tinib, whose clinical activity against NTRK fusion-positive cancers is not under investigation. Instead, ongoing trials involving cabo- zantinib, merestinib, altiratinib (DCC-22,701), and sitravatinib (MGCD-516), are being conducted either on unselected popula- tions, or populations selected for a broader range of oncogenic alterations that include NTRK fusions (NCT02920996, NCT01639508, NCT02228811, NCT02219711) [10].
Currently, two first-generation Trk inhibitors, entrectinib and larotrectinib, hold a therapeutic indication for NTRK fusion-positive cancers. The former is a potent Trk inhibitor whose inhibitory spectrum also encompasses the protein products of ALK and ROS1 fusions. The observed safety and

antitumor effectiveness (overall response rate [ORR] 57%- 63.5%) in phase-1 and phase-2 trials (ALKA-372-001, STARTRK-1, STARTRK-2) [26,27] led to the marketing authorization in US in August, 2019.
Other agents with limited inhibitory spectrum outside Trk have recently been synthesized. Those that reached clinical testing include taletrectinib (DS-6051b/AB-106) [28], belizatinib (TSR-011) [29], PLX-7486, repotrectinib (TPX-0005) [30], and selitrectinib (LOXO-195, BAY2731954) [31].

4.Larotrectinib
4.1.Chemistry
Larotrectinib (BAY2757556, formerly LOXO-101 and ARRY-470) (Box 1) is an orally bioavailable compound based on a 3-urea- substituted pyrazolo[1,5-a]pyrimidine structure. It was first synthesized by Array BioPharma (Boulder, Colorado, US) and first licensed to LOXO Oncology (Stamford, Connecticut, US) in 2013. Today, larotrectinib licensing rights are held by Bayer AG (Leverkusen, Germany).

4.2.Pharmacodynamics
Larotrectinib is a highly-selective pan-Trk small-molecule inhi- bitor, showing no significant off-target activity. Its mechanism of action relies on the competitive block of the ATP-binding site in the intracellular kinase domain of Trk receptors [32]. Its in vitro potency against Trk fusion proteins is characterized by IC50 values in the low nanomolar range [33]. In murine xeno- graft models, larotrectinib induced tumor regression at clini- cally achievable concentrations [33,34]. Larotrectinib activity is

limited to NTRK fusions, whereas tumors harboring NTRK
mutations did not show any response [15].

4.3.Pharmacokinetics
Larotrectinib is commercialized as 25 mg and 100 mg oral capsules, or as oral solution. The mean absolute bioavailability after a single dose of 100 mg was 34% (32% to 37%). The recommended dose is 100 mg b.i.d. for adults and 100 mg/m2
b.i.d. in the pediatric population, with a maximum dose of 100 mg b.i.d [15,35]. Plasma/tissue distribution is moderate (distribution volume 48 liters), and larotrectinib is 70% bound to plasma proteins, regardless of drug concentrations. The chief metabolization pathway is represented by hepatic cyto- chrome P450 (CYP) isoforms 3A4/5, mainly through O-glucuronidation. Coadministration with strong CYP isoform 3A4/5 inducers or inhibitors should be avoided: clinically rele- vant interactions can be predicted, as exposure to larotrectinib was altered by canonical metabolic modifiers as rifampine and itraconazole. Larotrectinib is also substrate for the efflux trans- porters P-glicoprotein and Breast Cancer Resistance Protein. The initial dose of larotrectinib should be reduced by 50% in patients with Child-Pugh B or C liver failure, whereas no dose modification is recommended in Child-Pugh A patients, or patients with renal failure [36]. The major route of excretion is biliary/fecal (58%), followed by renal/urine (39%). Larotrectinib is rapidly eliminated from plasma, with mean (standard deviation) half-life of 3.0 (1.5) hours at the recom- mended dose [15].

4.4.Clinical efficacy
4.4.1.Phase-1 and phase-2 studies
In the phase-1 dose-escalation LOXO-TRK-14001 trial (NCT02122913), larotrectinib was orally administered to 70 adult patients with metastatic solid tumors, regardless of NTRK fusion status. Different schedules of dosing in continu- ous 28-day cycles were tested in six cohorts (50, 100, 200 mg QD and 100, 150, 200 mg b.i.d.), to evaluate the safety profile and research dose-limiting toxicities (DLTs). Three DLTs were reported, each in a different cohort: consequently, the max- imum tolerated dose (MTD) was not reached, and, as men- tioned, 100 mg b.i.d. was the recommended dose for phase-2 patients, based on tolerability and antitumor activity observed [15].
The phase-1/2 SCOUT trial (NCT02637687) enrolled pedia-
tric and young patients with solid tumors, unselected for NTRK fusion status [35]. In the dose-escalation phase-1 part, 24 patients received larotrectinib b.i.d. in 28-day continuous cycles. Again, the MTD was not reached and, among the explored dosages, 100 mg/m2 b.i.d. (maximum 100 mg per dose) was recommended for the pediatric patients subse- quently enrolled in the phase-2 part, that evaluated antitumor activity. NTRK status emerged as a decisive predictor of response: a radiologic objective response was achieved in 14 (93%) of 15 patients with NTRK fusion-positive cancers, whereas the best response among all patients without NTRK fusions was progressive disease. The phase-2 part, as well as the follow-up of the phase-1 part, are still ongoing.

The ongoing NAVIGATE phase-2 trial (NCT02576431) enrolls adolescent and adult patients with NTRK fusion-positive can- cers. A pooled analysis of this and the aforementioned trials described safety and efficacy of larotrectinib in the first con- secutive 55 NTRK fusion-positive cancer patients [37]. Patients age ranged from 4 months to 76 years and cancer diagnoses spanned across 17 histologies. The ORR was 75% according to independent review and 80% by investigator assessment. Antitumor activity was observed regardless of age, tumor type, or NTRK fusion characteristics.
A subsequent pooled analysis of the three trials represents the largest published dataset of patients with NTRK fusion- positive cancers to date (159 patients) [38]. Median age was 43 years (range <1 month to 84 years); females were 52%; Eastern Cooperative Oncology Group performance status was 0 or 1 in 86%. The most frequent primary sites were: soft tissue sarcoma (44%; infantile fibrosarcoma 18%); thyroid (16%), sali-
vary gland (13%), lung (8%), colon (5%). Confirmed fusions
involved NRTK1 (40%), NTRK2 (3%), and NTRK3 (55%). After a median follow-up of 12.9 months, ORR among the 153 patients available for response evaluation was 79% (CI95% 72–85), and complete responses were observed in 16% of patients. Responses were observed across a wide range of primary sites, and were independent of the rearranged gene. Responses were also of rapid onset (median time to response
1.8 months), and long-lasting (median duration of response [DOR] 35.2 months). With a low number of survival events already occurred despite the longer follow-up (median [inter- quartile range] 13.9 months [6.5–24.9]), median progression- free survival and overall survival were 28.3 months (CI95% 22.1-not estimable; events 30%) and 44.4 months (CI95% 36.5-not estimable; events 14%), respectively. This high and durable activity translates into a meaningful growth modula- tion index [39]. A post-hoc analysis on small numbers sug- gested intracranial activity on brain metastases [38].

4.5.Safety and tolerability
The expanded analysis on the safety population (260 patients)
[38] confirmed the good tolerability observed in earlier experi- ences. Adverse events (AEs) were primarily of grade 1 or 2, with 46% of patients experiencing grade 3-4 AEs, the most common including anemia (10%), neutropenia (5%) and other laboratory abnormalities with individual incidence <5%. Furthermore, only 13.5% of patients experienced grade 3–4 AEs that were deemed related to treatment. While 5% of patients experienced one or more serious treatment-related AEs, no toxic deaths were observed. Dose reduction owing to AEs and treatment discontinuation were necessary only for 8% and 2% of patients, respectively.

4.6.Currently available trials
Currently, five phase-2 clinical trials with larotrectinib are recruiting pediatric, adolescent, and adult patients affected by NTRK fusion-positive solid or hematological cancers. Importantly, an expanded access program for larotrectinib is also available for all-age patients with NTRK fusion-positive

Table 2. Main primary sites that may feature NTRK fusions.
Clinicaltrials.gov identifier
Sponsor
Design
Diseases
Age Primary outcome (s)
NCT02637687 (SCOUT) Bayer Prospective, phase-2 part Solid tumors ≤21 y ORR
NCT02576431
(NAVIGATE) Bayer Prospective, phase-2 part Solid tumors Any ORR

NCT03834961 Children’s Oncology Group

Prospective, phase-2 Previously untreated solid tumors;
relapsed acute leukemia

≤30 y ORR

NCT03213704(Pediatric MATCH)
NCT03155620 (Pediatric MATCH)
NCT02465060(MATCH
Screening)

National Cancer Institute
National Cancer Institute
National Cancer Institute

Prospective, phase-2 Relapsed or refractory advanced solid
tumors, NHL, histiocytic disorders Prospective, phase-2 Relapsed or refractory advanced solid
tumors, NHL, histiocytic disorders Prospective, phase-2 Advanced refractory solid tumors,
lymphomas, MM

1-21 y ORR

1-21 y ORR

≥18 y ORR

NCT04655404 (CONNECT1903)

Nationwide Children’s Hospital

Prospective, early phase-1 High-grade glioma ≤21 y DCR, PK, PD, safety

NCT03025360 Bayer Expanded Access Any, ineligible for or cannot access
larotrectinib clinical trials

Any –

NCT04142437 (ON-TRK) Bayer Prospective, observational Any Any safety
Table 1. Ongoing clinical trials with larotrectinib in NTRK fusion-positive cancers (clinicaltrials.gov, www.umin.ac.jp) as of January, 2021. DCR, disease control rate; MM, multiple mieloma; NHL, non-Hodgkin lymphoma; ORR, overall response rate; PD, pharmacodynamics; PK, pharmacokinetics; y, years.

cancers who are ineligible for or cannot access an ongoing larotrectinib clinical trial (Table 2).

4.7.Regulatory affairs
On November 26th, 2018, the Food and Drug Administration (FDA) granted larotrectinib (VitrakviTM) accelerated approval in the United States for the treatment of adult and pediatric patients whose solid tumors harbor an NTRK fusion in the absence of a known resistance mutation, are metastatic (or surgical resection is likely to result in severe morbidity), and have no satisfactory alternative treatments or have progressed following treatment [36,40]. On July 26th, 2019, the Committee for Medicinal Products for Human Use of the European Medicines Agency recommended for a conditional marketing authorization in the European Union, which was granted on September 19th, 2019. Larotrectinib received approval in other countries, including Canada and Brazil, and is under evaluation in Japan [41].

5.Conclusion
NTRK fusions identify a unique molecular subgroup of solid tumors for which larotrectinib represents a highly effective and tolerable treatment. Regulatory approval rapidly followed the published promising results. Efforts should focus on how to better offer this option to the amenable general oncologic population.

6.Expert opinion
Historically, the development of targeted treatments for onco- genic driver-positive cancers followed the lane of the histol- ogy-specific trials. A new line of research then emerged, representing a paradigm shift in the advancement of precision oncology, more cognizant of the role played in tumorigenesis by these oncogenes across histologies. New basket trials were carried on, that would encompass different primary sites shar- ing a specific sensitizing mutation. Clinical research on NTRK fusions is fully inscribed in this philosophy, successfully

demonstrating an activity of pharmacological Trk inhibition regardless of the primary site. Trk inhibition emerges, thus, as a new histology-independent, biomarker-based treatment strategy. Indeed, larotrectinib was the second molecule to receive a tissue-agnostic FDA approval, the third being entrec- tinib one year later.
Following the regulatory approval of Trk inhibitors, NTRK fusion testing is already subject of recommendations from scientific societies [42–44]. While NTRK fusions are concen- trated in a handful of rare cancers, the bulk of cases amenable to treatment with larotrectinib is expected to come from the small share observed among the exceedingly more frequent malignancies, such as lung or colorectal cancer. This poses serious challenges regarding the incorporation of testing in the diagnostic work-up of these malignancies. Does the low incidence of the fusion warrant a routine blanket test? For instance, the benefit from target-therapies justified the com- prehensive search for ALK and ROS-1 rearrangements in lung cancer [22,45]; similarly, the presence of BRAFV600E point muta- tion provides useful clinical insights in colorectal cancer [46]. However, NTRK fusions in these common neoplasms are per- haps lower by one order of magnitude. Developing strategies to decrease the number-needed-to-screen is of paramount importance for the transfer of Trk inhibition into the daily oncological practice. NTRK fusions are mutually exclusive with commonly-researched oncogenic drivers (NRAS, KRAS, BRAF), and are associated with specific genomic characteristics (MLH1 promoter hypermethylation in colorectal cancer, low mutational burden in lung cancer). Ideally, a sequential approach should circumscribe by a large factor the population to be screened for NTRK fusions. On the other hand, this approach would exclude DNA mismatch-repair (MMR) profi- cient colorectal cancers, that may amount to half of cases harboring ALK, ROS1 or NTRK fusions [20]. Similarly, a selection of breast cancers according to the presence of a secretory phenotype, and of thyroid cancers according to the presence of papillary phenotype, may result in missing a quarter and a third of NTRK fusions, respectively [14,47].
Deeply intertwined is the issue of which technologies to employ in NTRK fusion detection. In low-prevalence

populations not requiring comprehensive genomic testing, an efficient, low-cost, easily-accessible screening can rely on pan- Trk immunohistochemistry (IHC) [48–50], albeit with limita- tions. Among them, reduced performance in some histologies, and inapplicability to central nervous system malignancies owing to physiologic expression of Trk proteins. In a two- stage approach, a positive IHC would require confirmation by next-generation sequencing (NGS), preferably RNA-based [48]. If a sequencing platform is available, upfront NGS is suggested as the preferred screening method, and IHC as confirmation of Trk protein expression. The role of fluorescent in situ hybridization is debated [11,44]. Against the backdrop of a single test (the NGS-based Foundation One CDX [Foundation Medicine Inc.]) FDA-approved as a companion diagnostic for larotrectinib [51], further research on screening algorithms and techniques is needed for an optimized NTRK fusions detection. For instance, the diagnostic application of antibodies directed against phosphorylated Trk proteins (e.g. ABN1381, PA5-36,695) may have a potential relevance in spe- cific conditions, in which a discrimination between Trk expres- sion and activation is necessary. This could be the case of the approach to central nervous system tumors, and the exclusion of a non-constitutively activated form in case of primary refractoriness.
Increased awareness of the biomarker, of its diagnostics, and of its therapeutic implications is essential, and will require diffuse education of oncologists and molecular pathologists, as well as the establishing -toward the era of molecular med- icine for all histotypes- of multidisciplinary molecular tumor boards.
A more proper definition of the safety profile will require longer follow-up. Particularly, although no indication of car- diac or neurologic toxicity emerged for larotrectinib so far, entrectinib-emergent neuropathy (resulting in dizziness, par- esthesias, weight gain, and cognitive disturbance) [26], along with manifestations in NTRK-deficient animal models (such as cardiac defects, loss of specific neuronal populations, memory loss, hyperphagia, and sympathetic neuropathy) [10] suggest caution when inferring long-term safety from the early evi- dence available. Hopefully, more mature data will dispel these concerns.
On the basis of the observed high response rates, further applications of Trk inhibition can be foreseen. First, the advancement of an efficacious and tolerable drug to the first line is highly desirable, however it will need to overcome the accrual and ethical challenges posed to randomized trials by the rarity of the genetic alteration. Second, Trk inhibition could find a neoadjuvant role in locally advanced or localized NRTK fusion-positive tumors, to facilitate margin-free resec- tions and spare mutilating or disfiguring surgery [52]. Third, the elevate cytocidal activity could very well translate into a massive release of neoantigens: upon these premises, the combination of larotrectinib with immunotherapy could be particularly relevant in colorectal cancer, in which the observed co-occurrence of NTRK fusions with MLH1 hyper- methylation mean that these tumors are also characterized by a MMR deficit. Trk inhibition could be used to enhance responses to anti-PD1 and anti-CTLA4 agents in this niche of patients [53,54], whose prognosis is likely to be negatively

impacted by NTRK fusions [21]. However, while providing a strong rationale for the combination of larotrectinib with immunotherapy, MMR deficiency could also represent an accelerating factor of the emergence of resistance to Trk inhibition, via the accumulation of a high mutational load. Fourth, TrkAIII-driven cancers, regardless of the presence of NTRK1 amplification, may also represent a further field of application for larotrectinib [16].
The evolution of first-generation Trk inhibitors is already a clinical need, as secondary drug resistance is increasingly being characterized. On-target resistance is driven by point mutations acquired in the Trk kinase domain regions of the solvent front, the activation loop xDFG motif, or at the gate- keeper position [37,55–57]. Importantly, second-generation Trk inhibitors proved able to overcome this resistance in vitro (ONO-5,390,556) [58], and also in early-phase clinical trials (selitrectinib, repotrectinib) [30,31,59–61]. Off-target resistance relies on aberrant oncogenic signaling coming from altered downstream Trk effectors, or from proteins implicated in other mitogenic, pro-survival pathways. Indeed, the specific inhibi- tion of aberrant BRAF or MET signaling successfully restored disease control in two Trk inhibitor-refractory NTRK fusion- positive tumors [62]. Albeit very preliminary, the global corpus of these data casts some light on the subsequent treatment of larotrectinib-refractory tumors, envisaging chemotherapy-free strategies for prolonging time under Trk inhibition, through agent sequencing in case of on-target resistance, or through the combination of specific inhibitors in case of off-target resistance.

Funding
This manuscript was not funded.

Declaration of interest
The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

ORCID
Roberto Filippi http://orcid.org/0000-0001-9698-7687 Ilaria Depetris http://orcid.org/0000-0001-6748-4992

References
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
1.Clary DO, Reichardt LF. An alternatively spliced form of the nerve growth factor receptor TrkA confers an enhanced response to neurotrophin 3. Proc Natl Acad Sci U S A. 1994;91 (23):11133–11137.

2.Ip NY, Stitt TN, Tapley P, et al. Similarities and differences in the way neurotrophins interact with the Trk receptors in neuronal and nonneuronal cells. Neuron. 1993;10(2):137–149.
3.Levi-Montalcini R. Morphological and metabolic effects of the nerve growth factor. Arch Biol (Liege). 1965;76(2):387–417.
4.Deinhardt K, Chao MV. Trk receptors. Handb Exp Pharmacol. 2014;220:103–119.
5.Eggert A, Sieverts H, Ikegaki N, et al. p75 mediated apoptosis in neuroblastoma cells is inhibited by expression of TrkA. Med Pediatr Oncol. 2000;35(6):573–576.
6.Nakagawara A. Trk receptor tyrosine kinases: a bridge between cancer and neural development. Cancer Lett. 2001;169(2):107–114.
7.Kummar S, Lassen UN, Inhibition TRK, et al. Treatment Strategy. Target Oncol. 2018;13(5):545–556.
8.Amatu A, Sartore-Bianchi A, Siena S. NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open. 2016;1(2):e000023.
9.Stransky N, Cerami E, Schalm S, et al. The landscape of kinase fusions in cancer. Nat Commun. 2014;5:4846.
10.Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol. 2018;15(12):731–747.
•• A comprehensive review of NTRK fusions biology and Trk inhibition mechanisms.
11.Penault-Llorca F, Rudzinski ER, Sepulveda AR. Testing algorithm for identification of patients with TRK fusion cancer. J Clin Pathol. 2019;72(7):460–467.
•This works provides useful insights on NTRK fusion testing.
12.Albert CM, Davis JL, Federman N, et al. TRK fusion cancers in children: a clinical review and recommendations for screening. J Clin Oncol. 2019;37(6):513–524.
13.Vaishnavi A, Le AT, Doebele RC. TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov. 2015;5(1):25–34.
14.Wilson TR, Sokol JS, Ross JS, et al. NTRK1/2/3 fusions in secretory versus non-secretory breast cancers. Ann Oncol. 2020;31: S292. Abstract #131P, ESMO Virtual Congress 2020.
15.Hong DS, Bauer TM, Lee JJ, et al. Larotrectinib in adult patients with solid tumours: a multi-centre, open-label, phase I dose-escalation study. Ann Oncol. 2019;30(2):325–331.
•Phase-1 trial evaluating larotrectinib in the adult population.
16.Farina AR, Cappabianca L, Ruggeri P, et al. The oncogenic neuro- trophin receptor tropomyosin-related kinase variant, TrkAIII. J Exp Clin Cancer Res. 2018;37(1):119.
17.Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364(26):2507–2516.
18.Sanz-Garcia E, Argiles G, Elez E, et al. BRAF mutant colorectal cancer: prognosis, treatment, and new perspectives. Ann Oncol. 2017;28(11):2648–2657.
19.Pietrantonio F, Petrelli F, Coinu A, et al. Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: a meta-analysis. Eur J Cancer. 2015;51(5):587–594.
20.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.
21.Wang J, Yi Y, Xiao Y, et al. Prevalence of recurrent oncogenic fusion in mismatch repair-deficient colorectal carcinoma with hyper- methylated MLH1 and wild-type BRAF and KRAS. Mod Pathol. 2019;32(7):1053–1064.
22.Solomon BJ, Kim DW, Wu YL, et al. Final overall survival analysis from a study comparing first-line crizotinib versus chemotherapy in ALK-mutation-positive non-small-cell lung cancer. J Clin Oncol. 2018;36(22):2251–2258.
23.Wirth LJ, Sherman E, Robinson B, et al. Efficacy of Selpercatinib in RET-altered thyroid cancers. N Engl J Med. 2020;383(9):825–835.
24.Kopetz S, Grothey A, Yaeger R, et al. Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. N Engl J Med. 2019;381(17):1632–1643.
25.Abou-Alfa GK, Sahai V, Hollebecque A, et al. Pemigatinib for pre- viously treated, locally advanced or metastatic

cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 2020;21(5):671–684.
26.Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: inte- grated analysis of three phase 1-2 trials. Lancet Oncol. 2020;21 (2):271–282.
•This work provides pooled data on entrectinib.
27.Rolfo CD, De Braud FG, Doebele RC, et al. Efficacy and safety of entrectinib in patients (pts) with NTRK-fusion positive (NTRK-fp) solid tumors: an updated integrated analysis. J clin oncol. 2020;38 (15_suppl):3605. Abstract #3605 2020 ASCO Annual Meeting.
28.Papadopoulos KP, Borazanci E, Shaw AT, et al. U.S. Phase I first-in- human study of taletrectinib (DS-6051b/AB-106), a ROS1/TRK inhi- bitor, in patients with advanced solid tumors. Clin Cancer Res. 2020;26(18):4785–4794.
29.Lin CC, Arkenau HT, Lu S, et al. A phase 1, open-label, dose-escalation trial of oral TSR-011 in patients with advanced solid tumours and lymphomas. Br J Cancer. 2019;121(2):131–138.
30.Drilon A, Ou SI, Cho BC, 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 (10):1227–1236.
31.Hyman D, Kummar S, Farago A, et al. Phase I and expanded access experience of LOXO-195 (BAY 2731954), a selective next-generation TRK inhibitor (TRKi). Cancer Res. 2019;79(13 Supplement):CT127. Abstract #CT127, AACR Annual Meeting 2019.
32.Vaishnavi A, Capelletti M, Le AT, et al. Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer. Nat Med. 2013;19(11):1469–1472.
33.Doebele RC, Davis LE, Vaishnavi A, et al. An oncogenic NTRK fusion in a patient with soft-tissue sarcoma with response to the Tropomyosin-Related Kinase inhibitor LOXO-101. Cancer Discov. 2015;5(10):1049–1057.
34.Hong DS, Brose MS, Doebele RC, et al. Clinical safety and activity from a phase 1 study of LOXO-101, a selective TRKA/B/C inhibi- tor, in solid-tumor patients with NTRK gene fusions. Mol Cancer Ther. 2015;14(12 Supplement 2):PR13. Abstract #PR13, AACR-NCI- EORTC International Conference: Molecular Targets and Cancer.
35.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.
•Phase-1 trial evaluating larotrectinib in the pediatric and ado- lescent population.
36.Larotrectinib US prescribing information 2018. cited 2020 Dec. Available at: www.accessdata.fda.gov/drugsatfda_docs/label/2018/ 211710s000lbl.pdf
37.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.
•Phase-2 trial evaluating larotrectinib activity.
38.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.
•• This work presents the largest dataset on larotrectinib avail- able in published literature.
39.Italiano A, Hong DS, Briggs A, et al. Growth modulation index (GMI) of larotrectinib versus prior systemic treatments for TRK fusion cancer patients. Ann Oncol. 2020;31(4 Supplement):S473–S474. Abstract #542P, ESMO Virtual Congress 2020.
40.FDA approves an oncology drug that targets a key genetic driver of cancer, rather than a specific type of tumor. Media release. cited 2020 Dec. Available at: www.fda.gov/news-events/press- announcements/fda-approves-oncology-drug-targets-key-genetic- driver-cancer-rather-specific-type-tumor
41.Bayer submits larotrectinib for marketing authorization in Japan for the treatment of TRK fusion cancer. Company media release. cited 2020 Dec. Available at: www.investor.bayer.com/en/nc/news/inves tor-news/investor-news/bayer-submits-larotrectinib-for-marketing- authorization-in-japan-for-the-treatment-of-trk-fusion-can/

42.Demetri GD, Antonescu CR, Bjerkehagen B, et al. Diagnosis and management of tropomyosin receptor kinase (TRK) fusion sarco- mas: expert recommendations from the World Sarcoma Network. Ann Oncol. 2020;11(31):1506–1517.
43.National Comprehensive Cancer Network. NCCN guidelines for non-small cell lung cancer (NSCLC). Version 8.2020.
44.Marchiò C, Scaltriti M, Ladanyi M, et al. ESMO recommendations on the standard methods to detect NTRK fusions in daily practice and clinical research. Ann Oncol. 2019;30(9):1417–1427.
•• ESMO recommendations on NRTK fusions detection: provides detailed insights on the available technologies.
45.Peters S, Camidge DR, Shaw AT, et al. Alectinib versus crizotinib in untreated ALK-Positive non-small-cell lung cancer. N Engl J Med. 2017;377(9):829–838.
46.Van Cutsem E, Cervantes A, Adam R, et al. ESMO consensus guide- lines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27(8):1386–1422.
47.Cabanillas ME, Drilon A, Farago AF, et al. Larotrectinib treatment of advanced TRK fusion thyroid cancer. Ann Oncol. 2020;31(4 Supplement):S1086. Abstract #1916P, ESMO Virtual Congress 2020.
48.Solomon JP, Linkov I, Rosado A, et al. NTRK fusion detection across multiple assays and 33,997 cases: diagnostic implications and pitfalls. Mod Pathol. 2020;33(1):38–46.
•This work presents detailed insights on NTRK detection.
49.Hechtman JF, Benayed R, Hyman DM, et al. Pan-Trk immunohisto- chemistry is an efficient and reliable screen for the detection of NTRK Fusions. Am J Surg Pathol. 2017;41(11):1547–1551.
50.Potts SJ, Dean EJ, Polikoff J, et al. Detecting NTRK, ROS1, and ALK gene fusions in gastrointestinal tumor patients. J Clin Oncol . 2017;35(4_suppl):619. Abstract #619, 2017 ASCO Gastrointestinal Cancers Symposium.
51.FDA approves Companion Diagnostic to identify NTRK fusions in solid tumors for larotrectinib. Media release. cited 2020 Dec. Available at: https://www.fda.gov/news-events/press- announcements/fda-approves-oncology-drug-targets-key-genetic- driver-cancer-rather-specific-type-tumor
52.DuBois SG, Laetsch TW, Federman N, et al. The use of neoadjuvant larotrectinib in the management of children with locally advanced TRK fusion sarcomas. Cancer. 2018;124(21):4241–4247.
53.
Andre T, Shiu K, Kim TW, et al. Pembrolizumab in microsatellite-instability-high advanced colorectal cancer. N Engl J Med. 2020 Dec 3;383(23):2207–2218.
54.Overman MJ, Lonardi S, Wong KYM, et al. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/ microsatellite instability-high metastatic colorectal cancer. J Clin Oncol. 2018;36(8):773–779.
55.Estrada-Bernal A, Le AT, Tuch B, et al. Identification of TRKA and TRKB kinase domain mutations that induce resistance to a pan-TRK inhibitor. Cancer Res. 2016;76(14 Supplement):LB–118. Abstract #LB-118, AACR Annual Meeting 2016.
56.Schram AM, Chang MT, Jonsson P, et al. Fusions in solid tumours: diagnostic strategies, targeted therapy, and acquired resistance. Nat Rev Clin Oncol. 2017;14(12):735–748.
57.Drilon A. TRK inhibitors in TRK fusion-positive cancers. Ann Oncol. 2019;30(Suppl_8):viii23–viii30.
58.Kozaki R, Yoshizawa T, Tsukamoto K, et al. A potent and selec- tive TRK inhibitor ONO-5390556, shows potent antitumor activ- ity against both TRK-rearranged cancers and the resistant mutants. Cancer Res. 2016;76(14 Supplement):2954A. Abstract #2954A, AACR Annual Meeting 2016.
59.Drilon A, Nagasubramanian R, Blake JF, 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(9):963–972.
60.Drilon A, Zhai D, Deng W, et al. Repotrectinib, a next generation TRK inhibitor, overcomes TRK resistance mutations including solvent front, gatekeeper and compound mutations. Cancer Res. 2019;79(13 Supplement):442. Abstract #442, AACR Annual Meeting 2016.
61.Drilon AE, Ignatius Ou SH, Cho BC, et al. A phase 1 study of the next-generation ALK/ROS1/TRK inhibitor ropotrectinib (TPX-0005) in patients with advanced ALK/ROS1/NTRK+ cancers (TRIDENT-1). J Clin Oncol. 2018;36(15_suppl):2513. Abstract #2513, 2018 ASCO Annual Meeting.
62.Cocco E, Schram AM, Kulick A, et al. Resistance to TRK inhibition mediated by convergent MAPK pathway activation. Nat Med. 2019;25(9):1422–1427.
•Valuable paper on the resistance to Trk first-generation inhibitors.Larotrectinib

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>