U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-.

Cover of StatPearls

StatPearls [Internet].

Show details

Tyrosine Kinase Inhibitors

; ; .

Author Information and Affiliations

Last Update: July 18, 2023.

Continuing Education Activity

Tyrosine kinase inhibitors (TKI) are a group of pharmacologic agents that disrupt the signal transduction pathways of protein kinases by several modes of inhibition. This activity will review the currently available drugs, their mechanism of action, routes of administration, indications, contraindications, and adverse effects.

Objectives:

  • Outline the different types of tyrosine kinase inhibitor (TKI) drugs and the currently available TKIs.
  • Describe the mechanism of action of tyrosine kinase inhibitors.
  • Summarize the indications and contraindications for each type of tyrosine kinase inhibitor.
  • Review the adverse effects and toxicity of tyrosine kinase inhibitors.
Access free multiple choice questions on this topic.

Indications

Mutations, dysregulation, and overexpression of protein kinases are involved in a multitude of disease processes. Around 1 in every 40 human genes codes for a protein kinase and nearly half of those genes map to either disease loci or cancer amplicons.[1] Interest in protein kinase inhibitors began with the FDA approval of the tyrosine kinase inhibitor (TKI) imatinib in 2001. Imatinib is an oral chemotherapy medication designed to target the BCR-Abl hybrid protein, a tyrosine kinase signaling protein produced in patients with Philadelphia-chromosome-positive chronic myelogenous leukemia. Since the introduction of Imatinib, the application of TKIs has been ever-expanding, particularly for cancer treatment, due to tyrosine kinases' critical roles in cellular signaling.[2][3]

Tyrosine kinase enzymes (TKs) can be categorized into receptor tyrosine kinases (RTKs), non-receptor tyrosine kinases (NRTKs), and a small group of dual-specificity kinases (DSK) which can phosphorylate serine, threonine, and tyrosine residues. RTKs are transmembrane receptor that includes vascular endothelial growth factor receptors (VEGFR), platelet-derived growth factor receptors (PDGFR), insulin receptor (InsR) family, and the ErbB receptor family, which includes epidermal growth factor receptors (EGFR) and the human epidermal growth factor receptor-2 (HER2). NRTKs are cytoplasmic proteins that consist of nine families, including Abl, Ack, Csk, Fak, Fes/Fer, Jak, Src, Syk/Zap70, and Tec, with the addition of Brl/Sik, Rak/Frk, Rlk/Txk, and Srm, which fall outside the nine defined families. The most notable example of DSKs is the mitogen-activated protein kinase kinases (MEKs), which are principally involved in the MAP pathways.[1][4][5]

As of now, there are over 50 FDA-approved TKIs. Comprehensive lists of FDA-approved TKIs with additional information are available at NIH PubChem and FDA.gov. Due to the broad reach of this topic and the rapid development of new drugs, this list is not fully comprehensive.

Image

Table

   TKI Target

Mechanism of Action

As a whole, tyrosine kinases phosphorylate specific amino acids on substrate enzymes, which subsequently alter signal transduction leading to downstream changes in cellular biology. The downstream signal transduction set off by TKs can modify cell growth, migration, differentiation, apoptosis, and death. Constitutive activation or inhibition, either by mutations or other means, can lead to dysregulated signal cascades, potentially resulting in malignancy and other pathologies.[4][40] Therefore, blocking these initial signals via TKIs can prevent the aberrant action of the mutated or dysfunctional TKs.

Despite the diverse primary amino acid sequences, human kinases share similar 3D structures, particularly when it comes to the ATP-binding pocket located in the catalytically active region. The starting amino acid sequence (ASP-Phe-Gly or DFG) of the flexible activation loop that controls access to the activation site is also typically conserved.[41]

Kinase inhibitors are either irreversible or reversible. The irreversible kinase inhibitors tend to covalently bind and block the ATP site resulting in irreversible inhibition. The reversible kinase inhibitors can further subdivide into four major subtypes based on the confirmation of the binding pocket as well as the DFG motif.[3][42]

Below are listed various binding modes of TKIs.[3]

  • Type I inhibitors: competitively bind to the ATP-binding site of active TKs. The arrangement of the DFG motif in type I inhibitors has the aspartate residue facing the catalytic site of the kinase.
  • Type II inhibitors: bind to inactive kinases, usually at the ATP-binding site. The DFG motif in type II inhibitors protrudes outward away from the ATP-binding site. Due to the outward rotation of the DFG motif, many type II inhibitors can also exploit regions adjacent to the ATP-binding site that would otherwise be inaccessible.
  • Type III inhibitors: do not interact with the ATP-binding pocket. Type III inhibitors exclusively bind to allosteric pockets adjacent to the ATP-binding region.
  • Type IV inhibitors: bind allosteric sites far removed from the ATP-binding pocket.
  • Type V inhibitors: refer to a proposed subset of kinase inhibitors that exhibit multiple binding modes.[43]

Administration

Nearly all TKIs are effective when taken orally. Therapeutic loading and maintenance dosages are unique to each drug and should require unique dosing for each patient. When administering specific TKIs, many factors can contribute to reduced potency and the development of acquired resistance. These factors include such as whether or not food intake affects bioavailability, the mechanism of drug metabolism and elimination, liver and kidney function, drug-drug interactions, the presence of other medications that alter stomach pH, and patient demographics.[44][45][46]

Adverse Effects

Adverse events of TKIs are usually dose-based, with broad side effect profiles unique to each drug. However, due to similarities in drug targets, different classes of TKIs can have similar side effect profiles. Clinicians use BCR-Abl and KIT inhibitors to treat Philadelphia chromosome-positive CML and GIST, respectively.[47] Both KIT and BCR-Abl inhibitors, Imatinib, in particular, are known to cause adverse cutaneous drug reactions.[48]

EGFRs are a large family of RTKs associated with several cancers, including NSCL, breast, colorectal, pancreatic, esophageal, and head-and-neck cancers. The most common severe adverse effects associated with EGRF inhibitors are related to cutaneous adverse drug reactions.[48][49] The reason for this association is likely due to EGFRs’ role in normal skin integrity. Inhibition of EGFR hinders integumental function leading to dysfunctional epidermal differentiation and re-epithelialization, resulting in skin erosions.[49]

Angiogenesis is a critical step in cancer growth. VEGF is a key inducer of pathological angiogenesis expressed in nearly all human tumors.[50] Due to VEGF’s role in blood vessel survival and plasticity, TKIs that inhibit VEGFR carry associations with several cardiovascular toxicities, particularly hypertension.[51][52] This toxicity is likely because VEGF is necessary for adequate nitric oxide production. Inhibition of VEGF, therefore, results in elevated systemic vascular resistance.[53][54] Because VEGF is vital to endothelial cell survival, anti-VEGF therapy can diminish the integrity and regenerative capacity of endothelial cells, causing pro-coagulant changes. The long-term weakening and diminished integrity of blood vessel walls can eventually lead to thrombosis and hemorrhage.[55] Other adverse events associated with VEGFR TKIs include: renal injury, left ventricular dysfunction, cerebral and intestinal hemorrhage, cardiac ischemia, thrombosis, and skin reactions.[38]

Common adverse events related to TKIs are listed below. Not all adverse events are associated with every TKI, occurring at different frequencies depending on the drug.

 General

  • Fatigue
  • Fevers/Chills
  • Weight loss/gain

Gastrointestinal

  • Abdominal pain
  • Diarrhea
  • Dysgeusia
  • Constipation
  • Nausea/vomiting
  • Hepatotoxicity
  • Stomatitis

Cardiovascular

  • Hypertension
  • Hypotension
  • Congestive heart failure
  • Thrombosis
  • Cardiac ischemia
  • Myocardial infarction
  • Strokes
  • Intestinal hemorrhage
  • QT interval prolongation

Dermatological

  • Steven Johnson syndrome
  • Toxic epidermal necrolysis
  • Drug rash with eosinophilia and systemic symptoms
  • Acute generalized exanthematous pustulosis
  • Palmar-plantar erythrodysesthesia
  • Maculopapular rashes
  • Cheilitis
  • Facial edema
  • Eczema
  • Pruritis
  • Photosensitivity
  • Xerosis
  • Alopecia
  • Hair color changes
  • Paronychia

Endocrine

  • Hypokalemia
  • Thyroid dysfunction
  • Lacrimation

Hematologic

  • Anemia
  • Thrombocytopenia
  • Neutropenia
  • Bruising

Musculoskeletal

  • Myalgias
  • Arthralgia

Ophthalmic

  • Central serous retinopathy
  • Retinal pigment epithelial detachment
  • TKI keratitis

Renal

  • Kidney dysfunction
  • Proteinuria

Respiratory

  • Interstitial pulmonary disease
  • Pneumonia
  • Upper respiratory tract infection
  • Dyspnea
  • Epistaxis
  • Rhinorrhea

Neurological

  • Cognitive impairment
  • Peripheral neuropathy
  • Headaches
  • Dizziness

Contraindications

There are very few contraindications for TKIs. Considering the use of TKIs in life-extending cancer therapy, the benefits associated with TKI use generally outweigh the risks.[56] Data concerning TKI use in pregnancy is sparse; however, with rising rates of advanced maternal-age pregnancies, cancers requiring TKI therapy during pregnancy have become more common.[57][58] While there are several reports of successfully administering TKIs, such as erlotinib, imatinib, and nilotinib, during pregnancy, multiple studies demonstrate adverse events and teratogenic effects related to TKI use during pregnancy.[36][59][60][61]

Data on TKI use during pregnancy is still lacking in most cases. As a result, TKIs are typically co-prescribed with an effective contraception method during therapy and several weeks after discontinuing the TKI. Other limitations to using TKIs include severe adverse reactions. Patients at particular risk for one of the known adverse effects of the TKIs about to be prescribed, such as hypertension, interstitial lung disease, and long-QT syndrome, should receive an alternative therapy if possible.

Monitoring

While TKIs relieve the disease burden of most cancer patients, acquired resistance through various mechanisms remains a bottleneck in cancer targeted therapy. Patients require monitoring for disease progression after the initial benefit, which could be a sign of acquired resistance.[46] Genetic testing to identify known resistance mutations can also help guide genotype-directed therapy.

Toxicity

TKIs are generally well-tolerated, especially when compared to non-targeted cancer therapy. However, only a few of these drugs are selective for only one target. Due to the ubiquitous physiological role protein kinases play in the body, toxicities affecting various organs can occur. Organs commonly affected include the heart, lungs, liver, gastrointestinal tract, kidneys, thyroid, blood, and skin.[62]

The toxicity and efficacy of TKIs are often closely linked; this allows on-target toxic effects to act as biomarkers of effective pharmacological inhibition for certain TKIs. For example, skin rashes can serve as a monitoring mechanism for the effects of some TKIs that target EGFR and hypertension and can generally help monitor the inhibition of VEGFR.[63][64][65] However, the combined detrimental effects of both on-target and off-target toxicities can diminish a patient’s quality of life and limit the dose intensity of their medication, leading to sub-therapeutic treatment.

The optimal TKI of choice and dose is a requirement to reduce toxicities and adverse events. While TIKs are mainly administered at a fixed dose, several factors must guide dosing when devising an optimal TKI regimen. These factors include drug-drug/drug-food interactions, genetic polymorphisms of ABC transporters, patient adherence, intestinal absorption, distribution, metabolism, and elimination.[66][67] The interplay of multiple processes regulating the pharmacokinetics and pharmacodynamics of TKIs merits consideration when administering a TKI to titrate the optimal dosage.[68]

Enhancing Healthcare Team Outcomes

The development of TKI represents one of the most significant medical breakthroughs of the 21st century; however, one of the drawbacks of this drug class, endemic to small molecule therapies for cancer treatment in general, is the financial burden to the patient.[1] Kinase inhibitor therapy ranges from $5000 to $10,000 per month or more in the United States.[68] The significant financial burden may contribute to non-compliance resulting in disease progression and treatment resistance.[69] Clinicians, pharmacists, and other healthcare professionals should be aware of these financial burdens and discuss them along with the other potential physical toxicities of these drugs with the patient.

Given the non-specific nature of this drug class, it is imperative that therapy is ordered and subsequently followed through the collaborative efforts of an interprofessional team. This interprofessional team includes clinicians (MDs, DOs, NPs, PAs), specialists, nursing staff, and pharmacists. Each team member needs to be knowledgeable about the indications, dosing, and potential adverse effects of these drugs so they can counsel the patient regarding expectations and monitoring for adverse events, which will allow any needed changes in therapy to be made promptly, thereby driving optimal patient therapeutic outcomes. [Level 5]

Review Questions

References

1.
Roskoski R. Properties of FDA-approved small molecule protein kinase inhibitors: A 2020 update. Pharmacol Res. 2020 Feb;152:104609. [PubMed: 31862477]
2.
Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002 Dec 06;298(5600):1912-34. [PubMed: 12471243]
3.
Wu P, Nielsen TE, Clausen MH. FDA-approved small-molecule kinase inhibitors. Trends Pharmacol Sci. 2015 Jul;36(7):422-39. [PubMed: 25975227]
4.
Jiao Q, Bi L, Ren Y, Song S, Wang Q, Wang YS. Advances in studies of tyrosine kinase inhibitors and their acquired resistance. Mol Cancer. 2018 Feb 19;17(1):36. [PMC free article: PMC5817861] [PubMed: 29455664]
5.
Patterson KI, Brummer T, O'Brien PM, Daly RJ. Dual-specificity phosphatases: critical regulators with diverse cellular targets. Biochem J. 2009 Mar 15;418(3):475-89. [PubMed: 19228121]
6.
Gebreyohannes YK, Wozniak A, Zhai ME, Wellens J, Cornillie J, Vanleeuw U, Evans E, Gardino AK, Lengauer C, Debiec-Rychter M, Sciot R, Schöffski P. Robust Activity of Avapritinib, Potent and Highly Selective Inhibitor of Mutated KIT, in Patient-derived Xenograft Models of Gastrointestinal Stromal Tumors. Clin Cancer Res. 2019 Jan 15;25(2):609-618. [PubMed: 30274985]
7.
Vansteenkiste JF, Van De Kerkhove C, Wauters E, Van Mol P. Capmatinib for the treatment of non-small cell lung cancer. Expert Rev Anticancer Ther. 2019 Aug;19(8):659-671. [PubMed: 31368815]
8.
Liu PCC, Koblish H, Wu L, Bowman K, Diamond S, DiMatteo D, Zhang Y, Hansbury M, Rupar M, Wen X, Collier P, Feldman P, Klabe R, Burke KA, Soloviev M, Gardiner C, He X, Volgina A, Covington M, Ruggeri B, Wynn R, Burn TC, Scherle P, Yeleswaram S, Yao W, Huber R, Hollis G. INCB054828 (pemigatinib), a potent and selective inhibitor of fibroblast growth factor receptors 1, 2, and 3, displays activity against genetically defined tumor models. PLoS One. 2020;15(4):e0231877. [PMC free article: PMC7313537] [PubMed: 32315352]
9.
Mazzocca A, Napolitano A, Silletta M, Spalato Ceruso M, Santini D, Tonini G, Vincenzi B. New frontiers in the medical management of gastrointestinal stromal tumours. Ther Adv Med Oncol. 2019;11:1758835919841946. [PMC free article: PMC6535752] [PubMed: 31205499]
10.
Markham A. Selpercatinib: First Approval. Drugs. 2020 Jul;80(11):1119-1124. [PMC free article: PMC7716849] [PubMed: 32557397]
11.
Dombi E, Baldwin A, Marcus LJ, Fisher MJ, Weiss B, Kim A, Whitcomb P, Martin S, Aschbacher-Smith LE, Rizvi TA, Wu J, Ershler R, Wolters P, Therrien J, Glod J, Belasco JB, Schorry E, Brofferio A, Starosta AJ, Gillespie A, Doyle AL, Ratner N, Widemann BC. Activity of Selumetinib in Neurofibromatosis Type 1-Related Plexiform Neurofibromas. N Engl J Med. 2016 Dec 29;375(26):2550-2560. [PMC free article: PMC5508592] [PubMed: 28029918]
12.
Lee A. Tucatinib: First Approval. Drugs. 2020 Jul;80(10):1033-1038. [PubMed: 32548668]
13.
Rolfo C, Ruiz R, Giovannetti E, Gil-Bazo I, Russo A, Passiglia F, Giallombardo M, Peeters M, Raez L. Entrectinib: a potent new TRK, ROS1, and ALK inhibitor. Expert Opin Investig Drugs. 2015;24(11):1493-500. [PubMed: 26457764]
14.
Marandino L, Raggi D, Giannatempo P, Farè E, Necchi A. Erdafitinib for the treatment of urothelial cancer. Expert Rev Anticancer Ther. 2019 Oct;19(10):835-846. [PubMed: 31544541]
15.
Bewersdorf JP, Jaszczur SM, Afifi S, Zhao JC, Zeidan AM. Beyond Ruxolitinib: Fedratinib and Other Emergent Treatment Options for Myelofibrosis. Cancer Manag Res. 2019;11:10777-10790. [PMC free article: PMC6935287] [PubMed: 31920387]
16.
Lamb YN. Pexidartinib: First Approval. Drugs. 2019 Nov;79(16):1805-1812. [PMC free article: PMC7044138] [PubMed: 31602563]
17.
Duggan S, Keam SJ. Upadacitinib: First Approval. Drugs. 2019 Nov;79(16):1819-1828. [PubMed: 31642025]
18.
Syed YY. Zanubrutinib: First Approval. Drugs. 2020 Jan;80(1):91-97. [PubMed: 31933167]
19.
Reddy V, Cohen S. JAK Inhibitors: What Is New? Curr Rheumatol Rep. 2020 Jul 22;22(9):50. [PubMed: 32700001]
20.
Queirolo P, Spagnolo F. Binimetinib for the treatment of NRAS-mutant melanoma. Expert Rev Anticancer Ther. 2017 Nov;17(11):985-990. [PubMed: 28851243]
21.
Shah RR, Shah DR. Safety and Tolerability of Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitors in Oncology. Drug Saf. 2019 Feb;42(2):181-198. [PubMed: 30649743]
22.
Newland A, Lee EJ, McDonald V, Bussel JB. Fostamatinib for persistent/chronic adult immune thrombocytopenia. Immunotherapy. 2018 Jan;10(1):9-25. [PubMed: 28967793]
23.
Zhao J, Song Y, Liu D. Gilteritinib: a novel FLT3 inhibitor for acute myeloid leukemia. Biomark Res. 2019;7:19. [PMC free article: PMC6737601] [PubMed: 31528345]
24.
Scott LJ. Larotrectinib: First Global Approval. Drugs. 2019 Feb;79(2):201-206. [PubMed: 30635837]
25.
Yang J, Gong W. Lorlatinib for the treatment of anaplastic lymphoma kinase-positive non-small cell lung cancer. Expert Rev Clin Pharmacol. 2019 Mar;12(3):173-178. [PubMed: 30657349]
26.
Liang C, Tian D, Ren X, Ding S, Jia M, Xin M, Thareja S. The development of Bruton's tyrosine kinase (BTK) inhibitors from 2012 to 2017: A mini-review. Eur J Med Chem. 2018 May 10;151:315-326. [PubMed: 29631132]
27.
Stone RM, Manley PW, Larson RA, Capdeville R. Midostaurin: its odyssey from discovery to approval for treating acute myeloid leukemia and advanced systemic mastocytosis. Blood Adv. 2018 Feb 27;2(4):444-453. [PMC free article: PMC5858474] [PubMed: 29487059]
28.
Paik J, Dhillon S. Alectinib: A Review in Advanced, ALK-Positive NSCLC. Drugs. 2018 Aug;78(12):1247-1257. [PubMed: 30030733]
29.
Signorelli J, Shah Gandhi A. Cobimetinib. Ann Pharmacother. 2017 Feb;51(2):146-153. [PubMed: 27701080]
30.
Cabanillas ME, Habra MA. Lenvatinib: Role in thyroid cancer and other solid tumors. Cancer Treat Rev. 2016 Jan;42:47-55. [PubMed: 26678514]
31.
Solassol I, Pinguet F, Quantin X. FDA- and EMA-Approved Tyrosine Kinase Inhibitors in Advanced EGFR-Mutated Non-Small Cell Lung Cancer: Safety, Tolerability, Plasma Concentration Monitoring, and Management. Biomolecules. 2019 Oct 30;9(11) [PMC free article: PMC6921037] [PubMed: 31671561]
32.
Shaw AT, Engelman JA. Ceritinib in ALK-rearranged non-small-cell lung cancer. N Engl J Med. 2014 Jun 26;370(26):2537-9. [PubMed: 24963575]
33.
Varone F, Sgalla G, Iovene B, Bruni T, Richeldi L. Nintedanib for the treatment of idiopathic pulmonary fibrosis. Expert Opin Pharmacother. 2018 Feb;19(2):167-175. [PubMed: 29327616]
34.
Zeiser R, Andrlová H, Meiss F. Trametinib (GSK1120212). Recent Results Cancer Res. 2018;211:91-100. [PubMed: 30069762]
35.
Bellesoeur A, Carton E, Alexandre J, Goldwasser F, Huillard O. Axitinib in the treatment of renal cell carcinoma: design, development, and place in therapy. Drug Des Devel Ther. 2017;11:2801-2811. [PMC free article: PMC5614734] [PubMed: 29033542]
36.
Pallera A, Altman JK, Berman E, Abboud CN, Bhatnagar B, Curtin P, DeAngelo DJ, Gotlib J, Hagelstrom RT, Hobbs G, Jagasia M, Kantarjian HM, Kropf P, Metheny L, Moore JO, Ontiveros E, Purev E, Quiery A, Reddy VV, Rose MG, Shah NP, Smith BD, Snyder DS, Sweet KL, Tibes R, Yang DT, Gregory K, Sundar H, Deininger M, Radich JP. NCCN Guidelines Insights: Chronic Myeloid Leukemia, Version 1.2017. J Natl Compr Canc Netw. 2016 Dec;14(12):1505-1512. [PubMed: 27956535]
37.
Jeong W, Doroshow JH, Kummar S. United States Food and Drug Administration approved oral kinase inhibitors for the treatment of malignancies. Curr Probl Cancer. 2013 May-Jun;37(3):110-44. [PMC free article: PMC3761410] [PubMed: 23972982]
38.
Lankhorst S, Danser AH, van den Meiracker AH. Endothelin-1 and antiangiogenesis. Am J Physiol Regul Integr Comp Physiol. 2016 Feb 01;310(3):R230-4. [PubMed: 26511523]
39.
Bhullar KS, Lagarón NO, McGowan EM, Parmar I, Jha A, Hubbard BP, Rupasinghe HPV. Kinase-targeted cancer therapies: progress, challenges and future directions. Mol Cancer. 2018 Feb 19;17(1):48. [PMC free article: PMC5817855] [PubMed: 29455673]
40.
Kolibaba KS, Druker BJ. Protein tyrosine kinases and cancer. Biochim Biophys Acta. 1997 Dec 09;1333(3):F217-48. [PubMed: 9426205]
41.
Tong M, Seeliger MA. Targeting conformational plasticity of protein kinases. ACS Chem Biol. 2015 Jan 16;10(1):190-200. [PubMed: 25486330]
42.
Norman RA, Toader D, Ferguson AD. Structural approaches to obtain kinase selectivity. Trends Pharmacol Sci. 2012 May;33(5):273-8. [PubMed: 22503441]
43.
Lamba V, Ghosh I. New directions in targeting protein kinases: focusing upon true allosteric and bivalent inhibitors. Curr Pharm Des. 2012;18(20):2936-45. [PubMed: 22571662]
44.
Wind S, Schnell D, Ebner T, Freiwald M, Stopfer P. Clinical Pharmacokinetics and Pharmacodynamics of Afatinib. Clin Pharmacokinet. 2017 Mar;56(3):235-250. [PMC free article: PMC5315738] [PubMed: 27470518]
45.
Kucharczuk CR, Ganetsky A, Vozniak JM. Drug-Drug Interactions, Safety, and Pharmacokinetics of EGFR Tyrosine Kinase Inhibitors for the Treatment of Non-Small Cell Lung Cancer. J Adv Pract Oncol. 2018 Mar;9(2):189-200. [PMC free article: PMC6302998] [PubMed: 30588353]
46.
Camidge DR, Pao W, Sequist LV. Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nat Rev Clin Oncol. 2014 Aug;11(8):473-81. [PubMed: 24981256]
47.
Jha P, Himanshu D, Jain N, Singh AK. Imatinib-induced Stevens-Johnsons syndrome. BMJ Case Rep. 2013 Jan 23;2013 [PMC free article: PMC3604560] [PubMed: 23349042]
48.
Ng CY, Chen CB, Wu MY, Wu J, Yang CH, Hui RC, Chang YC, Lu CW. Anticancer Drugs Induced Severe Adverse Cutaneous Drug Reactions: An Updated Review on the Risks Associated with Anticancer Targeted Therapy or Immunotherapies. J Immunol Res. 2018;2018:5376476. [PMC free article: PMC5822766] [PubMed: 29577050]
49.
Chen CB, Wu MY, Ng CY, Lu CW, Wu J, Kao PH, Yang CK, Peng MT, Huang CY, Chang WC, Hui RC, Yang CH, Yang SF, Chung WH, Su SC. Severe cutaneous adverse reactions induced by targeted anticancer therapies and immunotherapies. Cancer Manag Res. 2018;10:1259-1273. [PMC free article: PMC5962313] [PubMed: 29844705]
50.
Ferrara N. Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev. 2004 Aug;25(4):581-611. [PubMed: 15294883]
51.
Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M, Fahrig M, Vandenhoeck A, Harpal K, Eberhardt C, Declercq C, Pawling J, Moons L, Collen D, Risau W, Nagy A. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature. 1996 Apr 04;380(6573):435-9. [PubMed: 8602241]
52.
Kamba T, McDonald DM. Mechanisms of adverse effects of anti-VEGF therapy for cancer. Br J Cancer. 2007 Jun 18;96(12):1788-95. [PMC free article: PMC2359962] [PubMed: 17519900]
53.
Escalante CP, Zalpour A. Vascular endothelial growth factor inhibitor-induced hypertension: basics for primary care providers. Cardiol Res Pract. 2011;2011:816897. [PMC free article: PMC3099203] [PubMed: 21629798]
54.
Horowitz JR, Rivard A, van der Zee R, Hariawala M, Sheriff DD, Esakof DD, Chaudhry GM, Symes JF, Isner JM. Vascular endothelial growth factor/vascular permeability factor produces nitric oxide-dependent hypotension. Evidence for a maintenance role in quiescent adult endothelium. Arterioscler Thromb Vasc Biol. 1997 Nov;17(11):2793-800. [PubMed: 9409257]
55.
Kilickap S, Abali H, Celik I. Bevacizumab, bleeding, thrombosis, and warfarin. J Clin Oncol. 2003 Sep 15;21(18):3542; author reply 3543. [PubMed: 12972536]
56.
Rivas G, Llinás N, Bonilla C, Rubiano J, Cuello J, Arango N. Use of erlotinib throughout pregnancy: a case-report of a patient with metastatic lung adenocarcinoma. Lung Cancer. 2012 Aug;77(2):469-72. [PubMed: 22534670]
57.
Dou X, Qin Y, Huang X, Jiang Q. Planned Pregnancy in Female Patients with Chronic Myeloid Leukemia Receiving Tyrosine Kinase Inhibitor Therapy. Oncologist. 2019 Nov;24(11):e1141-e1147. [PMC free article: PMC6853119] [PubMed: 31186377]
58.
Ji Y, Schwartz J, Hartford A, Ramsey J, Phillips J, Verschraegen C. Successful Treatment of Non-Small Cell Lung Cancer With Erlotinib Throughout Pregnancy. JAMA Oncol. 2015 Sep;1(6):838-40. [PubMed: 26181671]
59.
Berman E, Druker BJ, Burwick R. Chronic Myelogenous Leukemia: Pregnancy in the Era of Stopping Tyrosine Kinase Inhibitor Therapy. J Clin Oncol. 2018 Apr 20;36(12):1250-1256. [PubMed: 29447062]
60.
Cortes JE, Abruzzese E, Chelysheva E, Guha M, Wallis N, Apperley JF. The impact of dasatinib on pregnancy outcomes. Am J Hematol. 2015 Dec;90(12):1111-5. [PMC free article: PMC5115878] [PubMed: 26348106]
61.
Pye SM, Cortes J, Ault P, Hatfield A, Kantarjian H, Pilot R, Rosti G, Apperley JF. The effects of imatinib on pregnancy outcome. Blood. 2008 Jun 15;111(12):5505-8. [PMC free article: PMC4916938] [PubMed: 18322153]
62.
Fujita KI, Ishida H, Kubota Y, Sasaki Y. Toxicities of Receptor Tyrosine Kinase Inhibitors in Cancer Pharmacotherapy: Management with Clinical Pharmacology. Curr Drug Metab. 2017;18(3):186-198. [PubMed: 28059038]
63.
Petrelli F, Borgonovo K, Cabiddu M, Lonati V, Barni S. Relationship between skin rash and outcome in non-small-cell lung cancer patients treated with anti-EGFR tyrosine kinase inhibitors: a literature-based meta-analysis of 24 trials. Lung Cancer. 2012 Oct;78(1):8-15. [PubMed: 22795701]
64.
Estfan B, Byrne M, Kim R. Sorafenib in advanced hepatocellular carcinoma: hypertension as a potential surrogate marker for efficacy. Am J Clin Oncol. 2013 Aug;36(4):319-24. [PubMed: 22547010]
65.
Hamnvik OP, Choueiri TK, Turchin A, McKay RR, Goyal L, Davis M, Kaymakcalan MD, Williams JS. Clinical risk factors for the development of hypertension in patients treated with inhibitors of the VEGF signaling pathway. Cancer. 2015 Jan 15;121(2):311-9. [PMC free article: PMC4293233] [PubMed: 25236375]
66.
Klümpen HJ, Samer CF, Mathijssen RH, Schellens JH, Gurney H. Moving towards dose individualization of tyrosine kinase inhibitors. Cancer Treat Rev. 2011 Jun;37(4):251-60. [PubMed: 20833478]
67.
Terada T, Noda S, Inui K. Management of dose variability and side effects for individualized cancer pharmacotherapy with tyrosine kinase inhibitors. Pharmacol Ther. 2015 Aug;152:125-34. [PubMed: 25976912]
68.
Roskoski R. Properties of FDA-approved small molecule protein kinase inhibitors. Pharmacol Res. 2019 Jun;144:19-50. [PubMed: 30877063]
69.
Dusetzina SB, Winn AN, Abel GA, Huskamp HA, Keating NL. Cost sharing and adherence to tyrosine kinase inhibitors for patients with chronic myeloid leukemia. J Clin Oncol. 2014 Feb 01;32(4):306-11. [PubMed: 24366936]

Disclosure: Robert Thomson declares no relevant financial relationships with ineligible companies.

Disclosure: Majid Moshirfar declares no relevant financial relationships with ineligible companies.

Disclosure: Yasmyne Ronquillo declares no relevant financial relationships with ineligible companies.

Copyright © 2024, StatPearls Publishing LLC.

This book is distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) ( http://creativecommons.org/licenses/by-nc-nd/4.0/ ), which permits others to distribute the work, provided that the article is not altered or used commercially. You are not required to obtain permission to distribute this article, provided that you credit the author and journal.

Bookshelf ID: NBK563322PMID: 33090752

Views

  • PubReader
  • Print View
  • Cite this Page

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...