Abstract
Molecular targeted tyrosine kinase inhibitors (TKIs) have produced unprecedented treatment response in cancer therapy for patients harboring specific oncogenic mutations. While the TKIs are mostly well tolerated, they were reported to increase serum levels of creatine kinase (CK) and cause muscle metabolism‐related toxicity. CK is an essential enzyme involved in cellular energy metabolism and muscle function. Elevated serum CK levels can arise from both physiological and pathological factors, as well as triggered by specific drug classes. The incidence of serum CK elevation induced by a few approved TKIs (brigatinib, binimetinib, cobimetinib‐vemurafenib combination [Food and Drug Administration, United States]; aumolertinib, and sunvozertinib [only approved by National Medical Products Administration, China]) were over 35%. CK elevation‐related symptoms include myopathy, myositis, inclusion body myositis (IBM), cardiotoxicity, rhabdomyolysis, rash, and acneiform dermatitis. High‐level or severe symptomatic CK elevation may necessitate dose reduction and indirectly dampen TKI efficacy. This review presents an updated summary about the prevalence rate and recent research about mechanisms leading to TKI‐induced serum CK elevation in cancer patients. The utility of monitoring serum CK levels for predicting TKI‐induced adverse effects and their management will also be discussed.
INTRODUCTION
Tyrosine kinases (TKs) are crucial components of cellular enzymatic machinery regulating signal transduction pathways, which play a pivotal role in cell proliferation, differentiation, apoptosis, and other biological processes. Dysregulation of TK‐regulated signaling is known to promote tumorigenesis. 1 Tyrosine kinase inhibitors (TKIs) are a large class of targeted therapeutic drugs that effectively suppress tumor growth and proliferation by inhibiting major oncogenic signaling pathways through the targeting of receptor or non‐receptor TKs. Over the past two decades, there has been significant progress in the development of TKIs targeting specific receptors including EGFR, VEGFR, ALK, ROS1, HER2, NTRK, RET, and MET. 1 The clinical application of these TKIs in chronic myeloid leukemia (CML), gastrointestinal stromal tumor (GIST), non‐small cell lung cancer (NSCLC), hepatocellular carcinoma (HCC), and other cancers, has significantly enhanced the survival and quality of life of patients. TKIs are generally well tolerated and they exhibit a more favorable toxicity profile than traditional chemotherapeutic drugs. Nevertheless, TKIs could still produce severe adverse events (AEs) such as cutaneous reaction, paronychia, and diarrhea. 2
Creatine kinase (CK), also known as creatine phosphokinase (CPK), is an important enzyme regulating cellular energy metabolism, muscle contraction, and adenosine triphosphate (ATP) regeneration. It catalyzes the reversible transfer of phosphate groups between creatine and ATP, thereby providing energy for muscle contraction and transport systems (Figure 1). It is usually present at low concentration in the bloodstream. Serum CK level can rise after heart attack, skeletal muscle injury, or strenuous exercise. There are three subtypes of CK, namely CK‐MM (predominantly found in skeletal muscles), CK‐MB (present in cardiac muscles), and CK‐BB (located in brain tissues). The measurement of CK in the blood has been applied in the diagnosis of conditions associated with muscle damage. To this end, the elevation of serum CK could also occur accompanying TKI‐induced adverse reactions. 1
FIGURE 1.
Schematic diagram illustrating the regulation of ATP production and consumption by the creatine kinase/phosphocreatine (CK/PCr) system. 3 The CK/PCr system is responsible for the temporal and spatial energy homeostasis in cells of high and fluctuating energy requirements. Creatine (Cr) enters the target cells via Cr transporter (CRT). Inside the cell, PCr/Cr and ATP/ADP equilibria are adjusted by a soluble fraction of cytosolic CK isoforms (CK‐c, see (3)). Another fraction of cytosolic CK (CK‐g, see (2)) is specifically coupled to glycolytic enzymes (G), accepting glycolytic ATP, while mitochondrial‐CK isoforms (mtCK, see (1)) is coupled to adenine nucleotide translocator (ANT), thus accepting ATP exported from the matrix and generated by oxidative phosphorylation (OP). The contribution of both of these microcompartments to the total PCr generation depends on the cell type. The PCr thus generated is fed into the large PCr pool (up to 30 mM) that is available as a temporal or spatial energy buffer. Another fraction of cytosolic CK (CK‐a, see (4)) specifically associated with subcellular sites of ATP utilization (ATPase; e.g., ATP‐dependent or ATP‐gated processes, ion‐pumps) also forms tightly coupled microcompartments regenerating the ATP utilized by the ATPase reaction in situ in the expense of PCr. The proposed CK/PCr energy shuttle or circuit connects the various subcellular sites of ATP production (glycolysis and mitochondrial oxidative phosphorylation) with subcellular sites of ATP utilization (ATPases), via highly diffusible PCr and Cr.
Elevated serum CK level is a prevalent clinical phenomenon, commonly observed due to various physiological factors, including age, gender (more obvious in males), race (more prevalent in individuals of African descent), geographical location, and strenuous exercise. 1 On the contrary, pathological elevation of CK level could indicate increased cell permeability or cellular damage in tissues containing CK, often resulting from neuromuscular disorders such as muscular dystrophy, metabolic myopathies, inflammatory myopathies, and central or peripheral nervous system disorders affecting the muscles. 1 Non‐neuromuscular conditions including cardiovascular disease, endocrine disorders, metabolic abnormalities, obstructive sleep apnea, connective tissue diseases, and malignant tumors can also lead to elevated serum CK levels. Interestingly, a few widely consumed medications, including statins (cholesterol‐lowering drugs), antipsychotics, antivirals, beta blockers, and colchicine alkaloids, are known to increase serum CK levels. 1
To this end, the incidence of CK elevation induced by a few TKIs (brigatinib, binimetinib, cobimetinib, and vemurafenib) has been reported to be more than 35%, which constituted the most common reasons for discontinuation or dose reduction. 4 , 5 , 6 Symptoms accompanied by CK elevation includes myopathy, 7 myalgia, 8 , 9 , 10 inclusion body myositis (IBM), 11 cardiotoxicity, 12 , 13 rhabdomyolysis, 14 , 15 , 16 , 17 and acneiform dermatitis. 18 High‐level or symptomatic CK elevation will affect patient adherence and indirectly affects the efficacy. Factors that influence the pharmacokinetics of TKIs and risk for TKI‐associated CK elevation including high‐dose TKI therapy, 19 polypharmacy, 20 , 21 , 22 drug–drug interactions (concomitant use of statins and TKI), 20 , 21 , 22 other co‐morbidities, 23 , 24 , 25 and pharmacogenetic considerations. 26 This review article provides an updated summary about the propensity of CK elevation in cancer patients receiving TKI therapy and its underlying mechanisms. The utility of monitoring CK elevation to predict TKI‐mediated adverse effects and drug efficacy will also be discussed.
PHYSIOLOGICAL FUNCTIONS OF CK AND ITS REGULATION
Normal physiological functions of CK
As depicted in Figure 1, CK regulates the transport of ATP/ADP into and out of the mitochondria, thereby controlling energy metabolism. 27 To date, at least five isoforms of CK have been identified: three isoenzymes in the cytoplasm (CK‐MM, CK‐MB, and CK‐BB) and another two isoenzymes (non‐sarcomeric and sarcomeric) in the mitochondria. 28 An elevated CK level in the bloodstream generally indicates the occurrence of or ongoing myocyte damage. 29 Following muscle cell damage, there is a significant accumulation of free fatty acids within the cells, resulting in alterations to membrane action potential thresholds and an increase in cell membrane permeability. This leads to the direct release of muscle enzymes into the bloodstream, causing elevated serum levels of CK. 30 Microscopic examination of muscle biopsy reveals varying sizes of muscle fibers accompanied by numerous rimmed vacuoles and abnormal proliferation of tubular structures, which may serve as the pathological basis for increased levels of muscle enzymes. 31
FACTORS LEADING TO SERUM CK ELEVATION
The mechanisms leading to CK elevation are multi‐factorial, which can be triggered by numerous physiological factors, systemic conditions, or various medications including TKIs. Physiological influences include race, gender, age, and exercise, with higher CK levels commonly seen in African and South Asian populations, males, and younger individuals 32 (Table 1). Systemic diseases such as malignant tumors, idiopathic inflammatory myopathies, hypertension, coronary heart disease, diabetes, and conditions such as rhabdomyolysis and hypothyroidism also contribute to raised CK levels. 34 Cachexia is present in up to 80% of patients with advanced cancer and in 60%–80% of those diagnosed with gastrointestinal, pancreatic, and lung cancers. 35 Yuko et al. compared muscle damage of cancer cachexia in mice bearing Lewis lung carcinoma/colon adenocarcinoma with that in a murine muscular dystrophy model (mdx mice). Unlike mdx mice, cachexia mice did not exhibit increase in serum CK level, and signs of regeneration were absent. In cancer cachexia, some humoral factors like TNFαand angiotensin II may block the regeneration process. 36 Medications, including statins, antipsychotics, and antiviral drugs, can lead to increased CK levels, which is often associated with muscular damage. 32
TABLE 1.
Factors contributing to serum creatine kinase (CK) elevation.
Factors | Detailed description |
---|---|
Non‐Caucasian ethnicity | Afro‐Caribbean ethnicity and muscular build 32 |
Strenuous exercise | CK level may increase within 24 h of strenuous physical activity 33 |
Endocrine disorders | Hypothyroidism, hyperparathyroidism 34 |
Electrolyte imbalance | Low sodium, potassium or phosphate 32 |
Drugs | Statins, fibrates, anti‐retrovirals, beta blockers, and TKIs 32 |
Muscular disorders | Inflammatory myopathies and sarcoid myopathy 34 |
Miscellaneous non‐muscular causes | Denervation, seizures, malignancy, and surgery 32 |
CK elevation caused by non‐oncology drugs
Medications are also known to be a significant and prevalent etiology for elevated serum CK levels. 1 Apart from anticancer drugs, various non‐oncology drugs such as statins, antipsychotics, antiviral drugs, beta blockers, colchicine, and donepezil have also been shown to increase serum CK levels. 37
Statins are notorious for inducing myalgia, muscle weakness, and rhabdomyolysis. 37 Approximately 5% of individuals on statin therapy may experience elevated levels of CK, typically ranging from 2 to 10 times the upper limit of normal (ULN). 38 In a prospective analysis of 2017 subjects at a single center, 1 171 exhibited serum CK elevation (>2 times the normal value). The implicated medications included statins (46.4%), fibrates (14.3%), antiretroviral drugs (14.3%), angiotensin II receptor antagonists (10.7%), immunosuppressants (7.1%), and hydroxychloroquine (7.1%).
Serum CK elevation caused by antipsychotic drugs is also relatively common. A recent systematic review 39 reported that the incidence rate of significant CK elevation (>10 times the ULN) after taking antipsychotic drugs ranged from 2% to 7%, albeit with 83% of the patients not experiencing any significant complications. Antiviral drugs are also known to induce serum CK levels. A cohort study 40 revealed that among 475 HIV‐1 infected patients treated with raltegravir (a HIV integrase inhibitor), 11.2% experienced elevated CK levels (≥3 times the ULN), resulting in a rate of 3.8 individuals per 100 people per year. Among these cases, only 1.5% of patients with elevated CK levels reported symptoms including muscle pain and/or contractures.
Serum CK elevation triggered by TKIs
The incidence of CK elevation induced by various classes of TKIs targeting ALK, MEK1/2, B‐Raf, FLT3, BCR‐Abl, EGFR, VEGFR, RET, KIT, and PDGFRα (Table 2) has been reported to be more than 20% in NSCLC, melanoma, ovarian cancer, colorectal cancer, AML, CML, RCC, and GIST. In particular, brigatinib, binimetinib, cobimetinib‐plus‐vemurafenib combination, and gilteritinib could induce serum CK level dramatically by over 35% (Table 3). Thus, CK elevation as a biomarker may reflect the complex interplay between the therapeutic targeting of TK pathways and the unintended interference with cellular functions beyond cancer cells. TKs are not only pivotal in the pathogenesis of various cancers but they also play substantial roles in normal cellular functions, including muscle metabolism. TKIs, while designed to inhibit the aberrant signaling in tumor cells, may inadvertently impact the normal activity of kinases within muscle tissue, leading to cellular stress or damage. This off‐target effect can lead to an increase in CK levels and muscle damage, which is clinically significant as it could herald the onset of adverse reactions such as myositis or cardiotoxicity. Monitoring CK levels in patients on TKIs becomes a crucial aspect of clinical management, allowing for the early detection and intervention of potential TKI‐induced muscle toxicity.
TABLE 2.
Small molecule tyrosine kinase inhibitors reported to induce serum creatine kinase (CK) elevation in clinical trials.
Primary targets | TKI | Company | Trade name | Year of FDA or NMPA approval | Therapeutic indications |
---|---|---|---|---|---|
ALK, ROS1 | Crizotinib | Pfizer | Xalkori | 2011 | ALK or ROS1‐positive NSCLC, inflammatory myofibroblastic tumors, anaplastic large cell lymphoma |
ALK, RET | Alectinib a | Roche | Alecensa | 2015 | ALK‐positive NSCLC |
ALK | Brigatinib a | Ariad Pharm | Alunbrig | 2017 | ALK‐positive NSCLC |
ALK, ROS1 | Iruplinalkib | Qilu | Qixinke | 2023 b | ALK‐positive NSCLC |
BCR‐ABL | Imatinib a | Novartis | Gleevec | 2001 | Ph + CML or ALL, aggressive systemic mastocytosis, chronic eosinophilic leukemia, dermatofibrosarcoma protuberans, hypereosinophilic syndrome, GIST, myelodysplastic/ myeloproliferative disease |
BCR‐ABL | Dasatinib |
Bristol Myers Squibb |
Sprycell | 2006 | Ph + CML or ALL |
BCR‐ABL | Bosutinib a | Pfizer | Bosulif | 2012 | Ph + CML |
BCR‐ABL | Asciminib a | Novartis | Scemblix | 2021 | Ph + CML |
B‐Raf | Vemurafenib a | Genentech | Zelboraf | 2011 | BRAFV600E melanomas, Erdheim‐Chester disease |
B‐Raf | Dabrafenib | GSK | Tafinlar | 2013 | BRAFV600E/K melanomas, BRAFV600E NSCLC, BRAFV600E anaplastic thyroid cancers |
B‐Raf | Encorafenib a |
Array BioPharma |
Braftovi | 2018 | Combination therapy with binimetinib for BRAFV600E/K melanomas |
CDK4/6 | Palbociclib | Parke‐Davis | Ibrance | 2015 | Estrogen receptor‐ and HER2‐positive breast cancers |
c‐MET | Merestinib | Lilly | — | Investigational | — |
EGFR | Gefitinib | AstraZeneca | Iressa | 2003 | NSCLC with exon 19 deletions or exon 21 substitutions |
EGFR | Osimertinib | AstraZeneca | Tagrisso | 2015 | NSCLC with exon 19 deletions or exon 21 substitutions, T790M resistance mutation |
EGFR | Aumolertinib a | Hansoh | Ameile | 2020 b | NSCLC with exon 19 deletions or exon 21 substitutions, T790M resistance mutation |
EGFR | Sunvozertinib | Dizal | Shuwozhe | 2023 b | NSCLC with ex20ins mutation |
EGFR | Oritinib | Sanhome | Shengruisha | Investigational | NSCLC with T790M resistance mutation |
FGFR2 | Futibatinib a | Tiaho Pharma | Lytgobi | 2022 | Bile duct cancers (cholangiocarcinomas) with FGFR2 fusions or other rearrangements |
FKBP12/mTOR | Everolimus | Novartis | Afinitor | 2009 | HER2‐negative breast cancer, pancreatic neuroendocrine tumors, RCC, angiomyolipomas, subependymal giant cell astrocytomas |
FLT3 | Gilteritinib a | Astellas Pharma | Xospata | 2018 | AML with FLT3 mutations |
KIT, PDGFRα | Ripretinib a |
Deciphera Pharma |
Qinlock | 2020 | Fourth‐line treatment for GIST |
MEK1/2 | Trametinib | GSK | Mekinist | 2013 | BRAFV600E/K melanoma, BRAFV600E NSCLC |
MEK1/2 | Cobimetinib a | Genentech | Cotellic | 2015 | BRAFV600E/K melanomas in combination with vemurafenib |
MEK1/2 | Binimetinib a |
Array BioPharma |
Mektovi | 2018 | Combination therapy with encorafenib for BRAFV600E/K melanomas |
MEK1/2 | Selumetinib | AstraZeneca | Koselugo | 2020 | Neurofibromatosis type I |
PDGFRα | Avapritinib |
Blueprint Medicines |
Ayvakit | 2020 | GIST with PDGFRα exon 18 mutations |
PI3K | Buparlisib | Novartis/Adlai Nortye | — | Investigational | — |
RET, VEGFR2 |
Cabozantinib | Exelixis | Cometriq | 2012 | Medullary thyroid cancer, RCC, HCC |
RET | BOS172738 | Daiichi Sankyo | — | Investigational | — |
VEGFR1/2/ 3 |
Sorafenib | Bayer | Nexavar | 2005 | HCC, RCC, differentiated thyroid cancer |
VEGFR2 | Sunitinib a | Pfizer | Sutent | 2006 | GIST, pancreatic neuroendocrine tumors, RCC |
VEGFR, RET | Lenvatinib a | Easai Co. | Lenvima | 2015 | Differentiated thyroid cancer |
Abbreviations: ALL, acute lymphoblastic leukemia; CML, chronic myeloid leukemia; GIST, gastrointestinal stromal tumor; HCC, hepatocellular carcinoma; NSCLC, non‐small cell lung cancer; RCC, renal cell carcinoma.
TKIs that result in more than 20% increase of CK levels in phase III trials with mono or combination therapy.
National Medical Products Administration (NMPA) approved.
TABLE 3.
Incidence of CK elevation by small molecule TKIs – their protein kinase targets and cancer type.
Primary targets | Reference/Trial name | TKI/drug combination | Tumor type | Trial type | N | All grade (%) a | ≥ grade 3 (%) a | Dose Reduction (%) | Symptoms |
---|---|---|---|---|---|---|---|---|---|
ALK | Camidge et al. 41 | Brigatinib | NSCLC (ALK TKI‐ naive) | Phase 3 | 136 | 50 | 26 | 18 b | Myalgia was reported in 10% patients; Musculoskeletal pain was reported in 11% patients. No grade 3 or higher myalgia or musculoskeletal pain was reported |
Gettinger et al. 19 | Brigatinib | NSCLC | Phase 1/2 | 79 | 16 | 5 | 0 | Neither myalgia nor musculoskeletal pain of any grade were reported in any patients | |
Gettinger et al. 19 | Brigatinib 90mg | NSCLC (progressed on crizotinib) | Phase 2 | 109 | 17 | 6 | 2 | Neither myalgia nor musculoskeletal pain of any grade were reported in any patients | |
Gettinger et al. 19 | Brigatinib 90 mg→180 mg | NSCLC (progressed on crizotinib) | Phase 2 | 110 | 34 | 14 | 9 | Neither myalgia nor musculoskeletal pain of any grade were reported in any patients | |
Yang et al. 5 (ALTA‐3) | Brigatinib | NSCLC (progressed on crizotinib) | Phase 3 | 125 | 70 | 36 | 11 | — | |
Yang et al. 5 | Alectinib | NSCLC | Phase 3 | 122 | 29 | 2 | 2 | — | |
Camidge et al. 41 | Crizotinib | NSCLC | Phase 3 | 137 | 17 | 1 | 2 | Myalgia was reported in 8% patients. Musculoskeletal pain was reported in 8% patients. No grade 3 or higher myalgia or musculoskeletal pain was reported | |
Shi et al. 42 | Iruplinalkib (WX‐0593) | NSCLC | Phase 2 | 146 | 34.9 | — | 2.1 | — | |
MEK1/2, B‐Raf | Dummer et al. 43 | Binimetinib and encorafenib | Melanoma | Phase 3 | 192 | 27.1 | 7.8 c | — | — |
Monk et al. 4 | Binimetinib | Ovarian carcinomas | Phase 3 | 200 | 50 | 26 | — | — | |
Tabernero et al. a 44 | Encorafenib, binimetinib, and cetuximab | Colorectal cancer | Phase 3 | 222 | 34.3 | 3 | — | — | |
Ascierto et al. 52 | Binimetinib | NRAS mutated melanoma | Phase 2 | 30 | 37 | 23 | 23 | Increase in serum CK level was the most common grade 3–4 TRAEs, though they were mostly asymptomatic. The most frequent symptoms of increased CK concentrations were muscle weakness in 4 patients and myalgia in 2 patients. | |
Ascierto et al. 52 | Binimetinib | Val600 BRAF mutated melanoma | Phase 2 | 41 | 22 | 17 | 12 | ||
Watanabe et al. 53 | Binimetinib | Solid tumors | Phase 1 | 21 | 81 | 33 | — | No clinical symptoms of myopathy were observed | |
Bardia et al. 54 | Binimetinib and buparlisib | Solid tumors | Phase 1b | 89 | 59.6 | 27 | — | — | |
Ascierto et al. 5 | Cobimetinib and Vemurafenib | Melanomas | Phase 3 | 247 | 35 | 12 | — | Increased serum CK level was generally asymptomatic and managed by close clinical observation | |
Dummer et al. 43 | Vemurafenib | Melanomas | Phase 3 | 186 | 2.2 | 0 | — | — | |
Ascierto et al. 5 | Vemurafenib and placebo | Melanomas | Phase 3 | 247 | 3 | <1 | — | — | |
Gross et al. 55 | Selumetinib | Neurofibromatosis | Phase 1/2 | 74 | 76 | 9 | 7 | The most common AE were asymptomatic CK. All participants who had a CK ≥ grade 2 (between 2.5 and 5 times above the upper limit of normal) had CK isoenzymes checked at the first occurrence, and no one had a concerning elevation in CK‐MB, indicating there was no significant cardiac component to the elevated CK | |
MEK1/2, PI3K | Bedard et al. 56 | Trametinib and buparlisib | RAS‐ or BRAF‐mutant non–small cell lung, ovarian, or pancreatic cancer | Phase 1b | 113 | 45.1 | 14.2 | — | — |
B‐Raf | Tabernero et al. 44 | Encorafenib and cetuximab | Colorectal cancer | Phase 3 | 216 | 3.7 | 0 | — | — |
Tabernero et al. 44 | Either cetuximab and irinotecan or cetuximab and FOLFIRI | Colorectal cancer | Phase 3 | 193 | 7.3 | 0.5 | — | — | |
BCR‐Abl | Adenis et al. 50 | Imatinib | GIST | Prospective | 87 | 45 | 0 | — | — |
Gordon et al. 16 | Imatinib | CML/GIST | Prospective | 25 | 80 | 8 | — | Of CK elevations patient, 3 had other medical conditions that could have contributed. 1 patient had a family history of myopathy, and 2 patients had a history of hypothyroidism although they were euthyroid during the study period. 1 patient had alcohol use of approximately 2 drinks nightly, and seven patients were on other medications associated with CK elevations (statins, antidepressants, and/or antipsychotics) | |
Gambacorti‐Passerini et al. 51 CTCAE 3.0 |
Imatinib | CML | Phase 3 | 251 | 20 | 5 | — | — | |
Bankar et al. 52 CTCAE 5.0 |
Imatinib | CML | Retrospective | 221 | 76 | — | — | Three patients experienced rhabdomyolysis while on treatment with imatinib and had to discontinue the treatment. | |
Franceschino et al. 53 | Imatinib | CML | Retrospective | 50 | 56 d | — | — | In a few cases (4%), treatment was interrupted as a precaution because of high (>1000) CK. No clinical signs of myopathy were noted and CK values returned within the normal range after imatinib discontinuation. Once imatinib was restarted, CK values tended to rise again. Myocardial‐CK (CK‐MB) values were evaluated in 6 patients with a large increase in total CK levels (>800) and found to be normal | |
Bankar et al. 52 | Dasatinib | CML | Retrospective | 31 | 65 | — | — | — | |
Bankar et al. 52 | Bosutinib | CML | Retrospective | 8 | 50 | — | — | — | |
Réa et al. 54 | Asciminib | CML | Phase 3 | 156 | 25 | 1.9 | — | — | |
Réa et al. 54 | Bosutinib | CML | Phase 3 | 76 | 23.7 | 5.3 | — | — | |
Gambacorti‐Passerini et al. 51 | Bosutinib | CML | Phase 3 | 248 | 8 | 1 | — | — | |
Bankar et al. 52 | Nilotinib | CML | Retrospective | 23 | 43 | — | — | — | |
Flt3 | Perl et al. 55 | Gilteritinib | AML | Phase 3 | 246 | 13.4 | 5.3 | — | — |
Usuki et al. 56 | Gilteritinib | AML | Phase 1 | 24 | 37.5 | 12.5 | — | — | |
EGFR | Lu et al. 57 | Aumolertinib | NSCLC | Phase 3 | 214 | 35.5 | 7 | 2.8 | No patient had a CK increase associated with rhabdomyolysis, and CK increase did not manifest as a serious event |
Malik et al. 58 | Osimertinib | NSCLC | Phase 4 | 60 | 10 | — | 3.3 | — | |
Parafianowicz et al. 10 | Osimertinib | NSCLC | Case series | 38 | 10 | 3 | — | 4 patients developed myositis | |
Xiong et al. 59 | Oritinib | NSCLC | Phase 2 | 286 | 28 | 4.5 | — | — | |
Wang et al. 60 | Sunvozertinib | NSCLC | Phase 2 | 104 | 57.7 | 17.3 | — | — | |
VEGFR, RET | Motzer et al. 61 | Sunitinib | RCC | Phase 3 | 375 | 49 | 3 | — | — |
Adenis et al. 50 | Sunitinib | Solid | Perspective | 155 | 29 | — | — | — | |
RET, VEGFR2 | Yamanaka et al. 62 | Cabozantinib | RCC | Retrospective | 13 | 61.5 | — | — | 2 patients with grade 2 or 3 of CK elevation developed rhabdomyolysis with muscle weakness and/or acute kidney injury |
Okubo et al. 63 | Cabozantinib | HCC | Prospective | 14 | 21 | 0 | — | The patients were without myalgia or muscle cramp | |
VEGFR1/2/3 | Adenis et al. 50 | Sorafenib | solid | Perspective | 155 | 19 | — | — | — |
FGFR2 | Goyal et al. 64 | Futibatinib | Cholangiocarcinoma | Phase 2 | 103 | 10 | 3 | — | — |
PDGFRα | Li et al. 65 | Avapritinib | GIST | Phase 1/2 | 65 | 46 | — | 8 | None of the events resulted in significant clinical symptoms |
c‐MET | Saleh et al. 66 | Merestinib and ramucirumab | Colorectal cancer | Phase 1 | 23 | 13 | 4.3 | — | — |
RET | Schoffski et al. 67 | BOS172738 | NSCLC and medullary thyroid cancers | Phase 1 | 67 | 54 | — | — | — |
Note: Blank entries: not mentioned.
Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; BID, twice daily; BIN, binimetinib; CML, chronic myeloid leukemia; ENC, encorafenib; GIST, gastrointestinal stromal tumor; HCC, hepatocellular carcinoma; HLH, hemophagocytic lymphohistiocytosis; IBM: inclusion body myositis; NSCLC, non‐small cell lung cancer; PEM, pembrolizumab; QD, once daily; RCC, renal cell carcinomas; TEAE, treatment‐emergent adverse events; TRAE, treatment‐related adverse events.
Grade 1: CK 1–2.5 ULN, Grade 2: 2.5–5 ULN, Grade 3: 5–10 ULN, and Grade 4: >10 ULN; Grade designation as per National Cancer Institute Common Terminology Criteria for Adverse Events, Version 3/4/4.03/5.
The initial protocol mandated dose interruption or reduction of brigatinib in the presence of grade 3 or 4 blood creatine phosphokinase level (CPK) elevations, irrespective of associated symptoms. In February 2021, the protocol was revised to require accompanying symptoms (grade 2 muscle pain or weakness) for dose modifications in cases of grade 3 or 4 CPK elevation.
In patients with BRAF mutation‐positive melanoma receiving binimetinib with encorafenib, rhabdomyolysis was reported in 1 patient (0.1%).
Elevated CK was defined as grade 2 or higher, showing higher than 620 U/L in men or 383 U/L in women during dasatinib treatment.
Incidence of serum CK elevation by different classes of TKIs
Anaplastic lymphoma kinase (ALK) inhibitors (brigatinib, alectinib, crizotinib, and iruplinalkib)
The incidence of all‐grade CK elevation by brigatinib and alectinib ranged from 16% to 70% and 10% to 29%. In phase III ALTA 1 L trial (NCT02737501), patients with ALK inhibitor‐naive advanced ALK‐positive NSCLC received brigatinib 180 mg/day (7‐day lead‐in at 90 mg once daily), and 50% of patients had CK elevation (Table 3). The incidence of Grade 3 or 4 CK elevation was 26%. Dose reduction for CK elevation occurred in 18% of patients. 41 In ALTA trial, brigatinib was applied to ALK‐positive NSCLC after disease progression on crizotinib, CK elevation occurred in 17% of patients in the 90 mg group and 34% of patients in the 90 → 180 mg group. The incidence of Grade 3–4 CK elevation was 6% in the 90 mg group and 14% in the 90 → 180 mg group. Dose reduction for CK elevation occurred in 2% of patients in the 90 mg group and 9% in the 90 → 180 mg group. 19 It seems to be dose‐dependent from 90 to 180 mg both in all grades and ≥Grade 3. In order to reduce the risk of early‐onset pulmonary events, the phase III ATLA‐3 trial introduces brigatinib at a lower dose before dose escalation. 5 , 68 In ATLA‐3, the incidence of treatment‐related CK elevation in ALK‐positive NSCLC patients on brigatinib or alectinib therapy was 70% and 29%, respectively. 5 In ALTA‐1 L, 17% of patients treated with crizotinib had increased CK. 41 The reasons for the differences in the clinical CK inhibitory profiles of different ALK inhibitors are not clear. A comparison between the ratio of steady‐state trough unbound drug concentration (Css, trough, unbound)/ALK inhibition constant (ALK Ki) was performed. The ratios were estimated from literature‐reported pharmacokinetics parameters and ALK Ki values of brigatinib (67 = 25 nM/0.37 nM), 69 , 70 alectinib (85 = 162 nM/1.9 nM), 71 , 72 and crizotinib (122 = 78 nM/0.64 nM), 73 , 74 respectively. Inhibition of the primary target (ALK) appears unlikely to explain the observed CK increases, as all three drugs are highly potent ALK inhibitors at their recommended doses, and there is no suggestion that the drug with the higher Css, trough, unbound/ALK Ki ratio at the therapeutic dosage was associated with greater CK increase. As such, it is possible that the differences may be explained by differential selectivity of the drugs and off‐target mechanisms or other factors, pointing to areas for future research.
MEK1/2, B‐Raf, PI3K inhibitors (binimetinib, encorafenib, cobimetinib, vemurafenib, selumetinib, dabrafenib, trametinib, and buparlisib)
The incidence of CK elevation induced by MEK1/2, B‐Raf, PI3K TKIs was generally high following monotherapy of binimetinib (22%–81%), 4 , 45 , 46 selumetinib (76%), 48 concurrent therapy of encorafenib and binimetinib (27.1%), 43 binimetinib‐plus‐buparlisib combination (59.6%), 47 buparlisib‐plus‐trametinib combination (45.1%), 49 vemurafenib‐plus‐cobimetinib combination (35%), 6 and triplet therapy of encorafenib, binimetinib and cetuximab (34.3%). 44 In Columbus study (NCT01909453), while encorafenib plus binimetinib showed favorable efficacy compared with vemurafenib, 27.1% of patients in combination group had increased CK and 7.8% were Grade 3–4, compared with 2.2% of all grades and none of Grade 3–4 in vemurafenib alone group. 43 Binimetinib was indicated for treatment of recurrent low‐grade serous ovarian carcinomas (LGSOCs; 30%–60% of these patients harbor KRAS/BRAF mutations) after at least one prior platinum‐based chemotherapy but less than three prior chemotherapeutic treatments. CK elevation was occurred in 50% of patients, 26% were ≥Grade 3. 4 In phase III BEACON study (NCT02928224), 665 patients with BRAF V600E mutated metastatic colorectal cancer were assigned to receive encorafenib, binimetinib, and cetuximab (triplet therapy); encorafenib and cetuximab (doublet‐therapy); or the investigators' choice of either cetuximab and irinotecan or cetuximab and FOLFIRI (folinic acid, fluorouracil, and irinotecan) (control group). Triplet therapy or doublet therapy trended to have improved OS, ORR, and progression‐free survival (PFS) compared with standard chemotherapy. CK elevation occurred in 34.3% in the triplet, 3.7% in doublet, and 7.3% in control group of all grades. 44 In two phase I/Ib studies, binimetinib alone or its combination with buparlisib (phosphatidylinositol3‐kinase [PI3K] inhibitor) were investigated in patients with advanced solid tumors bearing RAS/RAF alterations. Elevated CK levels were observed in 81% and 59.6% of patients in the two studies. 46 , 47
A phase III coBRIM study (NCT01689519) evaluated the combination of the BRAF inhibitor vemurafenib and the MEK inhibitor cobimetinib in previously untreated unresectable locally advanced or metastatic BRAF V600 mutation‐positive melanoma. 6 35% in combination group and 3% of patients in control group had increased CK. Elevated CK level of Grade 3 or worse occurred in 12% of 247 patients in the combination group versus one (<1%) of 246 in control group. 6
Of the 74 patients treated with selumetinib for neurofibromatosis type 11‐related plexiform neurofibromas, 76% of patients experienced serum CK elevation. 48 In a phase Ib dose‐escalation study, 113 patients with selected advanced solid tumors were enrolled to evaluate the combination of the pan‐PI3K inhibitor buparlisib (BKM120) and MEK1/2 inhibitor trametinib. Dose‐limiting toxicities (DLT) included stomatitis, diarrhea, dysphagia, and an increase in serum CK levels. 45.1% experienced elevated CK levels, with 14.2% as Grade 3–4. 49
BCR‐ABL (asciminib, bosutinib, dasatinib, and imatinib), FLT3 (gilteritinib), VEGFR1/2/3 inhibitors (sorafenib as multikinase inhibitor, sunitinib)
Serum CK elevation is frequently observed in patients treated with imatinib (20%–80%), 16 , 50 , 51 , 52 , 53 dasatinib (65%), 52 bosutinib (8%–50%), 51 , 52 , 54 asciminib (25%), 54 and gilteritinib (13.4%–37.5%). 55 , 56 Adenis et al. 50 conducted a prospective analysis on 155 eligible cancer patients from nine centers in France to assess the incidence of TKI‐induced CK elevation. GIST was identified as the most prevalent tumor type, accounting for 105 out of 155 cases (68%). Treatments were carried out with imatinib (56%), sunitinib (14%), sorafenib (10%), HER1/2 antagonists (erlotinib, gefitinib, and lapatinib, 9%), other TKIs (10%), and imatinib‐based combinations (1%). Myalgia was reported in 50/155 patients (32%). CK elevation was observed in 54 patients (35%). The incidence rates of CK elevation among patients treated with imatinib, sunitinib, sorafenib, HER1/2 antagonists, and other TKIs were found to be 45%, 29%, 19%, 14%, and 20%, respectively. Notably, patients who received any TKI treatment for more than 6 months demonstrated an increased likelihood of elevated CK levels. CK elevation occurred in 45% of 87 patients treated with imatinib in this study, while Franceschino et al. 53 reported a similar occurrence of CK elevation in 50 patients with CML receiving imatinib treatment. Considering that the patients included in Adenis's study had incomplete medication data at baseline and many experienced complications associated with concomitant medication usage, it is plausible that elevated CK levels could be attributed to their underlying medical conditions or other medications. 53
An updated analysis of the phase III BELA trial (NCT00574873) evaluated the safety profile and toxicity management of bosutinib compared with imatinib in adults with newly diagnosed chronic phase CML after more than 30 months from accrual completion. Among the 248 patients in the bosutinib group and 251 patients in the imatinib group who received at least one dose of treatment, there was a significantly lower incidence of CK elevation observed with bosutinib compared with imatinib (8% vs. 20%, p < 0.001). 51 When comparing the kinase inhibition profile of imatinib versus bosutinib, in addition to bcr‐abl TK inhibition, imatinib is also an inhibitor of the receptor TKs of platelet‐derived growth factor (PDGF) and stem cell factor (SCF), c‐kit. PDGF and its receptor (PDGFR) are integral to the pathophysiology of fiber regeneration, fibrosis, and the progression of muscle dystrophy, inhibition of which may potentially lead to direct myotoxic effects. In contrast, bosutinib does not inhibit c‐kit or PDGFR but predominantly targets the apoptosis‐linked STE20 kinases. This distinct kinase inhibition profile may contribute to the higher incidence of CK elevation by imatinib than bosutinib. 75
A phase III, open‐label, randomized study of asciminib, a BCR‐ABL1 inhibitor Specifically Targeting the ABL Myristoyl Pocket (STAMP), versus bosutinib in CML after two or more prior TKIs, the major molecular response (MMR) rate at week 24 was 25.5% with asciminib and 13.2% with bosutinib. 54 CK elevation was found in 25% in asciminib group and 23.7% in bosutinib group. 54 Gilteritinib is an oral, potent, selective FLT3 inhibitor with single‐agent activity in relapsed or refractory FLT3‐mutated AML; of the 246 patients in phase III study of gilteritinib, 13.4% of patients had increased CK, and 5.3% were Grade 3–5 including serious, life‐threatening, and fatal adverse reactions. 55
EGFR inhibitors (osimertinib, aumolertinib, oritinib, and sunvozertinib)
The incidence of serum CK elevation induced by EGFR‐TKIs (osimertinib, aumolertinib, oritinib, and sunvozertinib) is 10%, 10 , 58 35.5%, 57 28%, 59 and 57.7%, 60 respectively. The third‐generation EGFR‐TKIs osimertinib, aumolertinib, and oritinib are oral and irreversible third‐generation EGFR‐TKIs selectively targeting both sensitizing EGFR and EGFR T790M mutations. 59 , 76 , 77 In AENEAS phase III study (NCT03849768), the most common AEs in the aumolertinib group was CK increase with 35% of all grade and 7% ≥Grade 3. 57 In the four major clinical studies of osimertinib: AURA (NCT01802632), 78 AURA2 (NCT02094261), 53 AURA3 (NCT02151981), 79 and FLAURA (NCT02296125), 80 CK elevation was only reported in AURA2, none of which explicitly reported the occurrence of adverse effects such as myositis, myalgia, or rhabdomyolysis. But fatigue, weakness, and elevated liver enzymes may be manifestations of muscle damage and cause CK elevation as reported in the above study. In a study conducted in India assessing the safety of osimertinib in patients with stage IV NSCLC with EGFR T790M mutation‐positive, CK increase (10%) was one of the most frequently observed treatment‐emergent adverse events (TEAEs). Parafianowicz et al. 81 reported that myositis accompanied by elevated CK occurred in 4 out of 38 patients receiving osimertinib (10%).
A phase II study assessing SH‐1028 (oritinib) in EGFR T790M‐positive advanced NSCLC showed that the incidence of treatment‐related CK elevation was 28% (n = 286), and the incidence of CK elevation ≥Grade 3 was 4.5%. 59 Sunvozertinib (DZD9008) is a selective, irreversible EGFR exon20 insertion inhibitor. In phase II WU‐KONG6 study, 104 patients received 300 mg sunvozertinib, 57.7% experienced CK elevation, and 17.3% were ≥Grade 3. 60
FGFR2, VEGFR, RET, FKBP12/mTOR, VEGFR2 inhibitors (futibatinib, lenvatinib, sunitinib, everolimus, and cabozantinib)
The incidence of CK elevation induced by sunitinib, cabozantinib, futibatinib is 29%–49%, 50 , 61 21%–61.5%, 62 , 63 and 10%, 64 respectively. A randomized phase III trial (NCT00083889) demonstrated superiority of sunitinib over interferon alfa in PFS as first‐line treatment for metastatic renal cell carcinoma (RCC), based on CTCAE 3.0, 49% of patients in sunitinib group displayed CK elevation. 61
A retrospective study of 13 patients with advanced RCC who were treated with cabozantinib monotherapy. Eight (61.5%) experienced an increase in serum CK levels, with five patients classified as Grade 1 elevation. The elevation of CK occurred at a median of 14 days after the initiation of cabozantinib treatment. Rhabdomyolysis, accompanied by muscle weakness and/or acute kidney injury, developed in two patients who experienced Grade 2 or 3 CK elevation. 62 In TAS‐120‐101 study (NCT02052778), futibatinib (a fibroblast growth factor receptor (FGFR) 1–4 inhibitor) was investigated in FGFR2 fusion‐positive or FGFR2 rearrangement‐positive intrahepatic cholangiocarcinoma. Ten percent of the patients exhibited serum CK elevation, among whom 3% were considered ≥Grade 3. 64
KIT, PDGFRα, c‐MET, and RET inhibitors (ripretinib, avapritinib, BOS172738, and merestinib)
The incidence of CK elevation induced by ripretinib, avapritinib, BOS172738, and merestinib plus ramucirumab combination was 21%, 46%, 65 54%, 67 and 13%, 66 respectively. Avapritinib is a type 1 kinase inhibitor designed to potently and selectively inhibit oncogenic KIT/PDGFRA mutants by targeting the kinase active conformation. A phase I/II bridging study (NCT04254939) evaluating the safety and the antineoplastic activity of avapritinib in Chinese patients with unresectable/metastatic GISTs (N = 65); 30 patients had elevated CK (46%), and five patients were ≥Grade 3 (8%). 65 In a phase Ia/b study (NCT02745769), patients were treated with combination of the type II MET kinase inhibitor, merestinib, and the antivascular endothelial growth factor receptor 2 monoclonal antibody, ramucirumab; CK elevation was occurred in 13% of patients. 66 BOS172738 is a selective oral RET kinase inhibitor designed with nanomolar potency against RET and >300‐fold selectivity against vascular endothelial growth factor receptor 2, in phase I study (NCT03780517); 54% of patients had CK elevation among 67 patients with RET‐altered advanced solid tumors. 67
Clinical symptoms / adverse events associated with TKI‐induced CK elevation
Myopathy, myalgia, weakness, and IBM
The most common symptom associated with serum CK elevation is myopathy. A myopathy is a disorder characterized by a primary functional or structural impairment of the skeletal muscles. Myopathy is presented with (1) Proximal symmetric weakness, which may manifest acutely, subacutely, or chronically. (2) Muscle bulk that is reduced, preserved, or enlarged. (3) Muscle pain or discomfort upon palpation (myalgia). (4) Muscle stiffness or cramps. (5) Asthenia (weakness) and fatigue. (6) Myoglobinuria (the presence of myoglobin in the urine). 81 In a phase II study (NCT01320085), patients with NRAS‐mutated or BRAF‐mutated advanced melanoma receiving at least one dose of binimetinib (n = 71) were investigated. Twenty patients displayed CK elevation (28.2%) and their most common symptoms were muscle weakness (4 out of 20 patients) and myalgia (2 out of 20 patients). 73 Patients with myopathy are more likely to develop elevated CK. Adenis et al. investigated serum CK increase in 155 solid tumor patients treated with TKI. The presence of spontaneous myalgias was significantly associated with abnormal CK levels (26/50, 52% in case of myalgias vs. 28/105, 27% in the absence of myalgias, chi‐squared test: p = 0.002). 50
There are notable case reports of myopathy accompanied by significant CK elevation induced by osimertinib, alectinib, and dasatinib, specific TKI targeting EGFR, ALK, and BCR/ABL, respectively. In a few clinical studies, NSCLC patients treated with osimertinib developed severe leg spasms, myalgia, or myositis, which were associated with a significant elevation of serum CK level from 989 U/L to 29,680 U/L. 7 , 9 , 82 A patient with ALK‐rearranged lung cancer treated with alectinib after a temporary drug halt due to elevated CK levels of 2673 U/L; the treatment was successfully resumed without further myositis episodes. 83 Moreover, a 69‐year‐old man with imatinib‐refractory CML developed progressive muscle weakness and IBM following treatment with dasatinib, which was characterized by a significant rise in serum CK levels (>Grade 3). 11 Due to the promising dasatinib‐induced tumor remission in this patient, dasatinib therapy was resumed under close clinical monitoring.
Cardiotoxicity
TKI‐induced serum CK elevation is generally related to skeletal muscle damage. In the case of TKI‐cardiotoxicity, the related rise of serum CK (particularly, the CK‐MB isotype preferentially expressed in the heart) may be utilized as a potential biomarker for monitoring. Electrocardiogram (ECG) screening is recommended for patients at risk for a thorough evaluation of their cardiovascular condition prior to initiating TKI therapy, in order to mitigate the potential cardiotoxic effects such as arrhythmias, acute coronary syndrome (ACS), congestive heart failure, and pleural effusion. 84 Interestingly, Al‐Ali et al. 85 investigated 113 patients treated with imatinib for at least 6 months. CK electrophoresis revealed elevated CK‐MM in 83% (including one patient with Makro‐CK‐I) and elevated CK‐MB in 17%. Therefore, identifying the specific subtype of elevated CK may be critical in determining the presence of myocardial damage following TKI treatment.
Pechbach et al. reported an asymptomatic and reversible ventricular arrhythmia induced by dasatinib in a 54‐year‐old patient on second‐line treatment for the management of chronic phase CML. Dasatinib was temporarily discontinued for 1 week to ensure complete systemic elimination and then restarted and associated with an anti‐arrhythmic regimen of metoprolol and flecainide. The patient developed proximal limb myalgias following the initial dose. A 24‐h ECG was repeated, revealing a restoration to normal sinus rhythm. The patient experienced a good clinical and paraclinical response following homograft. 12 The combination of TKI and immune checkpoint inhibitors may be associated with more toxicities. Guo et al. reported two cases having myocarditis due to cobimetinib and atezolizumab treatment both received long‐term pembrolizumab before disease progression. One patient with CK elevated to 1165 U/L and high‐sensitivity troponin T (hs‐TnT) at 77 ng/L. Transthoracic echocardiography (TTE) revealed a left ventricular ejection fraction (LVEF) of 55%. Cardiac magnetic resonance imaging (CMRI) demonstrated midwall myocardial edema and late gadolinium enhancement (LGE) in the basal, anterolateral, mid‐inferior, and inferior segments consistent with myocardial inflammation and necrosis. The other patient was asymptomatic but had an elevated CK level of 630 U/L and hs‐TnT level of 455 ng/L. Both patients received methylprednisolone and corticosteroids which resulted in decreased levels of CK and hs‐TnT, respectively. However, one patient experienced disease progression 4.6 months after the diagnosis of myocarditis, while the other patient did not respond to alternative ipilimumab‐plus‐nivolumab therapy and complications related to immune‐related nephritis requiring prednisolone. 13
Rhabdomyolysis
Rhabdomyolysis is characterized by progressive proximal muscle weakness, general weakness, and muscle pain, especially in the lower extremities, and brown‐colored urine. 86 In laboratory work‐up, clinicians commonly use serum CK levels that exceed five times the ULN value to diagnose rhabdomyolysis. 86 Severe symptomatic rhabdomyolysis is a rare AE reported in a few pivotal clinical studies investigating TKIs. However, monotherapy of a number of TKIs (including cabozantinib, cobimetinib, erlotinib, dasatinib, trametinib, imatinib, sunitinib, sorafenib, and osimertinib), combination therapy of binimetinib with encorafenib, dabrafenib and trametinib, ribociclib and simvastatin, pazopanib and rosuvastatin, ribociclib and statin, and triplet therapy of palbociclib, fulvestrant, and statin (atorvastatin), palbociclib, fulvestrant, and statin (simvastatin) have been reported to induce rhabdomyolysis. The severity of CK elevation varied widely among patients, from >5 times upper normal limit (UNL) to levels exceeding 10 times UNL, encompassing a range from reversible symptoms to life‐threatening cases of rhabdomyolysis. In the coBRIM trial (NCT01689519), Grade 3 or 4 CK elevations, including asymptomatic elevations over baseline, occurred in 12% of patients receiving cobimetinib with vemurafenib and 0.4% of patients receiving vemurafenib. The median time to first occurrence of Grade 3 or 4 CK elevations was 16 days in patients receiving cobimetinib with vemurafenib; the median time to complete resolution was 15 days. Elevation of serum CK increase of more than 10 times the baseline value with a concurrent increase in serum creatinine of 1.5 times or greater compared with baseline occurred in 3.6% of patients receiving cobimetinib with vemurafenib and in 0.4% of patients receiving vemurafenib. 6 In the COLUMBUS trial (NCT01909453), rhabdomyolysis was reported following combination therapy with binimetinib and encorafenib. Elevation of serum CK was also reported in 58% of patients treated with binimetinib‐encorafenib combination. In patients with BRAF mutation‐positive melanoma receiving binimetinib plus encorafenib (n = 690), rhabdomyolysis was reported in one patient (0.1%). 87
Two patients showed a more than 50% reduction in left ventricular ejection fraction and massive elevation of CK, which was accompanied by myoglobinuria and renal failure. 60 These two cases may be particularly severe because both patients were nephrectomized and their conditions were complicated by multiorgan failure. A retrospective study of 13 advanced RCC patients treated with cabozantinib monotherapy. Eight patients (61.5%) experienced an increase in serum CK levels, among which two patients with Grade 2 or 3 CK elevation developed rhabdomyolysis characterized by muscle weakness and/or acute kidney injury. 62
There were a few clinical reports about the potential risk of rhabdomyolysis resulting from drug–drug interactions between TKIs and statins. It has been reported that patients with RCC and advanced breast cancer could experience severe muscle toxicity following pazopanib, ribociclib, or palbociclib therapy if the patients were also on statins (such as rosuvastatin, simvastatin, and atorvastatin). 20 , 21 , 22 , 26 , 88 Statins, while effective at lowering cholesterol, have a rare but serious side effect of muscle injury, which can escalate to rhabdomyolysis and potentially leading to kidney damage. The interaction is exacerbated by the inhibition of CYP3A4, an enzyme responsible for metabolizing some certain statins like simvastatin, rosuvastatin, and atorvastatin, by some TKIs (e.g., palbociclib and ribociclib) and a potential inhibitor of the uptake transporter OATP1B1 of statin. 26 The reports highlighted cases with CK levels ranging from 3070 to 37,000 U/L and even a fatal outcome with levels at 14,572 U/L. 20 , 21 , 22 , 26 , 88 Serious symptoms associated with rhabdomyolysis caused by statins and TKIs include secondary acute kidney injury, 21 tetraparesis, 26 and necrotizing rhabdomyolysis related to death. 22 These interactions suggest that clinicians should be vigilant in monitoring CK levels when prescribing combinations of these medications. The genetic predisposition of the patients should also be considered, which could affect drug metabolism.
Other symptoms or adverse events
Elevated serum CK levels were also associated with skin rash induced by antineoplastic drugs. Garcia et al. 89 investigated the levels of CK in 287 patients on TKI therapy (targeting EGFR/HER2, m‐TOR, VEGFR, SRC/ABL, Aurora kinase, BRAF/MEK, PARP, CDK, or A5B1 integrin). Patients with Grade 2/3 rash were found to display the highest incidence of serum CK elevation (67%) compared with those with no or Grade 1 rash. Following resolution of the rash, a significant decrease in CK values was observed in 25 systematically monitored patients (8.7%) (p = 0.012). In vitro exposure of human keratinocytes (HaCaT) to EGFR, MEK, and PI3 kinase/m‐TOR inhibitors resulted in upregulated expression of CK‐B but not CK‐M or mitochondrial‐CK. 89 Binimetinib treatment in two melanoma patients was reported to trigger varying AEs accompanied by serum CK elevation. One patient experienced severe myalgia, Grade 3 CK elevation, Grade 2 acneiform dermatitis, bilateral retinopathy, and increased eye pressure. The other patient suffered from Grade 4 CK elevation and Grade 2 facial acneiform dermatitis without myalgia. 18
Management of adverse effect associated with CK elevation after TKI therapy
A representative algorithm for dose adjustment following CK elevation has been proposed based on the clinically approved United States prescribing recommendation of various TKIs. Appropriate dose adjustment is recommended according to the severity of serum CK elevation (Grade 1: CK 1–2.5 times ULN; Grade 2: 2.5–5 times ULN; Grade 3: 5–10 times ULN; and Grade 4 > 10 times ULN) (Table 4). Briefly, if CK elevation is considered Grade 0–2, there is no need to interrupt the treatment or adjust the dose. For some TKIs (brigatinib, binimetinib, and gilteritinib), serum CK assessment is recommended prior to treatment. If the extent of CK elevation is higher than or equal to Grade 3, the TKI therapy should be suspended for a period until the situation is improved to Grade 1/2 or baseline. Afterward, the TKI therapy may be resumed in full dose or at a reduced dose. If there is no improvement in serum CK level following the treatment suspension, the TKI therapy should be permanently discontinued.
TABLE 4.
Representative algorithm for treatment interruption and dose reduction of tyrosine kinase inhibitor (TKI) therapy as recommended by the approved United States Prescribing Information of various TKIs. Appropriate dose adjustment is recommended according to the severity of serum CK elevation.
Drug | Grade or severity of CK elevation a | Grade or severity of muscle symptom b | Treatment | Dose adjustment recommendation |
---|---|---|---|---|
All TKI | Grade 0–2 | — | Unaltered | No modification |
Grade 3 or 4 | Grade 2 or higher symptoms |
First recurrence: try to restart at full dose Second recurrence: reduce the dose at the next lower dose If recurrence after 60 mg, the drug should be permanently discontinued |
||
Cobimetinib 90 | Grade 4 | — | Suspend therapy for up to 4 weeks |
If improved to Grade 3 or lower, resume at the next lower dose level If not improved within 4 weeks, permanently discontinue |
Any grade | Myalgia | |||
Binimetinib 91 | Grade 4 asymptomatic | — | Suspend therapy for up to 4 weeks |
If improved to Grade 0–1 resume at a reduced dose If not resolved within 4 weeks, permanently discontinue |
Any grade | With symptoms or with renal impairment | |||
Encorafenib 92 | Grade 3 | — | Suspend therapy for up to 4 weeks |
If improves to Grade 0–1 or to pretreatment/baseline level, resume at reduced dose If no improvement, permanently discontinue |
— | Grade 3 | |||
Grade 4 | — | Suspend therapy for up to 4 weeks or permanently treatment discontinuation |
If improves to Grade 0–1 or to pretreatment/baseline level, resume at reduced dose If no improvement, permanent treatment discontinuation is recommended |
|
— | Grade 4 | |||
Gilteritinib 93 | ≥Grade 3 | — | Interrupt until toxicity resolves or improves to Grade 1 | Resume TKI therapy at 80 mg |
— | ≥Grade 3 |
Grade 1: CK 1–2.5 times ULN; Grade 2: 2.5–5 times ULN; Grade 3: 5–10 times ULN; and Grade 4: >10 times ULN; Grading is defined according to National Cancer Institute Common Terminology Criteria for Adverse Events. Version 4.0 or 4.03.
Muscle symptoms including muscle pain or weakness.
Table 5 summarized representative clinical cases of serum CK elevation which is accompanied by myopathy, myositis, IBM, ventricular arrhythmia, myocarditis, rhabdomyolysis, acneiform dermatitis, and other TKI‐induced symptoms. In most situations, after excluding the possibility of CK elevation caused by other confounding factors (comorbidities and concurrent medications), temporary suspension or dose reduction of TKI was shown to ameliorate the clinical symptoms. Some cases revealed that dose reduction of TKI could resolve the symptoms while still maintaining tumor suppression (Table 5). In NSCLC patients on osimertinib therapy, the clinical symptoms (including myositis) associated with elevated serum CK could be relieved by employing a two consecutive days on and 1 day off regimen, approximately 53 mg/day, which is higher than half dose (40 mg/day). 81 In another case report of NSCLC patient who developed myositis following osimertinib therapy, dose reduction by half after a one‐month osimeritinib suspension was found to resolve the symptoms despite the persistent elevation of CK above the normal range. 9 On the contrary, in melanoma patient treated with binimetinib, treatment suspension, and subsequent dose reduction was adopted. 18 Upon resumption of binimetinib therapy, the same AEs recurred, another dose reduction of 2‐weeks‐on and 1‐week‐off schedule with reduced dose (30 mg b.i.d.) was commenced. With this dose reduction regimen, CK elevation was maintained between Grade 1 and Grade 3. Importantly, the severe AEs including acneiform dermatitis and retinopathy were mostly resolved and an effective suppression of tumor burden was observed. 18 Intermittent dosing of MEK inhibitors has been proposed as a potential strategy to prevent drug resistance in melanoma. The underlying hypothesis is that the on‐treatment periods target drug‐sensitive tumor cells, while the off‐treatment intervals drive drug‐resistant cells into cell cycle arrest and apoptosis. Early treatment with MEK inhibitors shows higher tumor immunogenicity, indicating a strong antitumor immune response. However, prolonged exposure can trigger a phenotypic switch in melanoma cells, leading to increased invasiveness and reduced expression of melanocytic differentiation markers in MITF‐expressing cells. 101 There is study reporting a surge of T cells in the first week, followed by a decline, suggesting reduced immunogenicity over time. 102 Choi et al. 103 found that pulsatile treatment better maintains T‐cell activity and extends survival in KRAS‐mutant cancers. In another case report of melanoma patient experiencing trametinib‐induced rhabdomyolysis, a reduced dose of dabrafenib (200 mg/day) and trametinib (1.5 mg/day) was reported to allow the normalization of serum CK level and resolution of rhabdomyolysis. Upon further follow‐up, the patient developed muscle weakness with a slightly elevated serum CK level. A further dose reduction to dabrafenib (150 mg/day) and trametinib (1 mg/day) was recommended, when the patient was still free from rhabdomyolysis. Importantly, both primary and metastatic tumor lesions of the patient were under control even on the reduced doses of dabrafenib and trametinib for at least 6 months. 98
TABLE 5.
Management and follow‐up treatments of cancer patients experiencing serum creatine kinase (CK) elevation, who were accompanied by myopathy, myositis, IBM, ventricular arrhythmia, myocarditis, rhabdomyolysis, acneiform dermatitis, and other tyrosine kinase inhibitor (TKI)‐induced symptoms.
TKI | Target | Cancer type | Symptom(s) | CK value (U/L) | Management | Resolution of adverse effects after intervention | Follow‐up anticancer treatment |
---|---|---|---|---|---|---|---|
Osimeritinib 7 | EGFR | NSCLC | Myopathy | 989 | Discontinuation | CK level improved but higher than normal | Osimertinib |
Gefitinib 8 | EGFR | NSCLC | Myositis | Grade 2 | Discontinuation | Muscle symptoms disappeared with CK normalization | — |
Osimertinib 9 | EGFR | NSCLC | Myositis | 1238 | Discontinuation | Myalgia subsided and CK levels decreased | Half‐dose osimertinib (40 mg) |
Osimertinib 10 | EGFR | NSCLC | Myositis | 790 | Discontinuation | CK normalization | 2 days on and 1 day off |
Osimertinib 10 | EGFR | NSCLC | Myositis | 2511 | Discontinuation | CK normalization | 2 days on and 1 day off |
Osimertinib 10 | EGFR | NSCLC | Myositis | 596 | Discontinuation | CK normalization | 2 days on and 1 day off |
Osimertinib 10 | EGFR | NSCLC | Myositis | 298 | Discontinuation | CK normalization | 2 days on and 1 day off |
Osimertinib 82 | EGFR | NSCLC | Myositis, hepatitis, presynaptic muscle fiber damage caused by profound muscle necrosis | 29,680 | Discontinuation; Symptom management: prednisone (40 mg orally daily), pulse‐dose steroids Solu‐Medrol 60 mg intravenous (IV) daily and IV immunoglobulin 1 g/kg daily for 2 days, pyridostigmine 2 mg IV | Distal strength recovery, proximal muscle weakness, able to eat a regular diet and did not require supplemental oxygen and transferred to acute rehabilitation | Chemotherapy and stereotactic brain radiation |
Alectinib 83 | ALK, RET | Lung cancer with BM | Myositis | 2673 | Discontinuation | — | Alectinib |
Dasatinib 11 | BCR‐Abl | CML | IBM | >Grade 3 | TKI therapy was not discontinued because of complete molecular remission by dasatinib and the patient did not receive any additional treatment | CK levels remain unchanged, clinical status has not worsened, and he remains with levels of BCR‐ABL indicating a minimum of molecular response 4.5 | Dasatinib |
Dasatinib 12 | BCR‐Abl | CML | Ventricular arrhythmia | Elevated CK with myalgias | Suspended for 7 days | Electrocardiogram reading returned to baseline | Dasatinib, metoprolol, and flecainide |
Cobimetinib and atezolizumab 13 | MEK1/2 | Melanoma | Myocarditis | 1165 | Methylprednisolone | CK and hs‐TnT reduced, preserved LVEF and reduced midwall edema | — |
Cobimetinib and atezolizumab 13 | MEK1/2 | Melanoma | Myocarditis | 377 | Methylprednisolone | CK and hs‐TnT normalization | Ipilimumab and nivolumab (ineffective) |
Erlotinib 14 | EGFR | NSCLC | Rhabdomyolysis | Grade 3 |
Discontinuation; Symptom management: hydration and intravenous bicarbonate infusion |
All vital signs returned to the normal ranges, reduction of myalgia | — |
Dasatinib 15 | BCR‐Abl | Ph + CML | Rhabdomyolysis | 3831 | Dasatinib was stopped and the patient was given aggressive hydration, allopurinol, and diuretics | — | — |
Imatinib 16 | BCR‐Abl | CML | Rhabdomyolysis | Discontinuation, intravenous hydration, steroids | Normalization of CK | Dose reduction of imatinib | |
Imatinib 17 | BCR‐Abl | Fibromatosis (desmoid tumors) | Rhabdomyolysis | 1068 | Discontinuation | Normalization of CK | Radiotherapy |
Sunitinib a , 94 | VEGFR2 | RCC | Rhabdomyolysis | 3849 | — | Patient succumbed | — |
Sunitinib a , b , 94 | VEGFR2 | RCC | Rhabdomyolysis | 3160 | Hemodialysis | Recovered and all laboratory tests normalized | — |
Sorafenib 95 | VEGFR1/2/3 | HCC | Rhabdomyolysis | 10,911 |
Discontinuation; Symptom management: intravenous fluids |
Myalgia and fatigue resolved | Best supportive care |
Osimertinib 96 | EGFR | NSCLC | Rhabdomyolysis, acute renal insufficiency, hyperuricemia, metabolic acidosis, and electrolyte disturbances | 1470 |
Osimertinib discontinuation; Symptom management: alkalinization of urine and rehydration |
All symptoms improved | — |
Dabrafenib and trametinib 97 | B‐Raf, MEK1/2 | Melanoma | Hemophagocytic lymphohistiocytosis (HLH), rhabdomyolysis | 10 times UNL | Treatment discontinuation with basic supportive care (intravenous rehydration and paracetamol) with no requirement for steroids | The patient's condition rapidly improved and EBV viral load was negative in blood after 3 weeks | Vemurafenib/cobimetinib combination |
Dabrafenib and trametinib 97 | B‐Raf, MEK1/2 | Melanoma | Hemophagocytic lymphohistiocytosis (HLH), rhabdomyolysis | 5 times UNL | Treatment discontinuation with supportive care measures (intravenous rehydration and paracetamol) and oral steroids (1 mg/kg/day) | Condition rapidly improved | Encorafenib/binimetinib combination (including steroid in the regimen) |
Trametinib 98 | MEK1/2 | Melanoma | Rhabdomyolysis | 7312 | Treatment discontinuation with intravenous fluids administration | Normalization of CK | Dabrafenib 200 → trametinib (1.5 mg) and dabrafenib (200 mg) |
Dabrafenib and trametinib 99 | B‐Raf, MEK1/2 | Melanoma | Rhabdomyolysis | 14,328 | i.v. prednisolone 40 mg/day with fluid replacement | CK level (186 IU/L) and myoglobin (45 ng/mL) decreased | Nivolumab (2 mg/kg every 3 weeks) |
Dabrafenib and trametinib 100 | B‐Raf, MEK1/2 | Melanoma | Rhabdomyolysis, renal failure, and visual loss | 2218 | Discontinuation | Most of the side effects resolved within 3 weeks, but some retinal lesions and the abduction deficit persisted | — |
Pazopanib and rosuvastatin 20 | VEGFR1/2/3 | RCC | Rhabdomyolysis | 5719 | Discontinuation of both drugs, fluid resuscitation | A full recovery | — |
Ribociclib and statin 21 | CDK4/6 | Breast cancer | Rhabdomyolysis with secondary AKI | 3070 | Discontinuation of simvastatin and ribociclib | Condition gradually improved, creatinine gradually decreased to 1.3 mg/dL, GFR to 40.2 mL/min/1.73 m2 | Ribociclib 200 mg, simvastatin was permanently discontinued |
Palbociclib, fulvestrant, and statin (atorvastatin) 22 | CDK4/6 | Breast cancer | Necrotizing rhabdomyolysis | 14,572 | Atorvastatin, palbociclib, and fulvestrant were discontinued, hydration, methylprednisolone | Adverse effects continued to deteriorate; patient succumbed | — |
Palbociclib, fulvestrant, and statin (simvastatin) 26 | CDK4/6 | Breast cancer | Tetraparesis and rhabdomyolysis | >22,000 | Atorvastatin, palbociclib, and fulvestrant were discontinued, hydrated with 4 L saline i.v., intravenous immunoglobulin (IVIG) 0,4 g/kg | Gait function restored after 2 weeks of hospitalization and no signs of neuromuscular deficiency after 1‐year follow‐up | — |
Ribociclib and simvastatin 88 | CDK4/6 | Breast cancer | Rhabdomyolysis |
CK: 37,000 U/L, creatinine: 105 μmol/L Grade 3 neutropenia AST/ALT: 1314/377 UI/L |
Discontinuation of ribociclib and simvastatin, rehydration was initiated with 2 L of NaCl 0.9% and 1 L of bicarbonate, ICU, intensified hydration with 2 L of NaCl 0.9% for the first 5 h in association with 3 L of glucose 5% on 24 h, intravenous nicardipine |
Normalization of blood tests: CK: 102 U/L, creatinine: 63 mmol/L, AST/ALT: 36/77 UI/L |
Ribociclib 200 → 400 → 600 mg/day, with good tolerance, and continued 600 mg/day for 3 weeks followed by 7 day off Simvastatin was stopped |
Binimetinib 18 | MEK1/2 | Melanoma | Acneiform dermatitis G2, bilateral retinopathy G1, and eye pressure elevation G2. | Grade 3 | Discontinuation | All AEs resolved | Dose reduction of binimetinib (30 mg b.i.d.) and intermittent treatment (3 weeks on and 10 days off) |
Binimetinib 18 | MEK1/2 | Melanoma | Facial acneiform dermatitis G2 and retinopathy G1, but no myalgias | Grade 4 | 2‐week‐on and 1‐week‐off schedule with reduced dose (30 mg b.i.d.) | CK elevation was between G1 and G3, acneiform dermatitis between G1 and G2, and the retinopathy between G0 and G1 | 2‐weeks‐on and 1‐week‐off schedule with reduced dose (30 mg b.i.d.) |
Abbreviations: AKI, acute kidney injury; BM, brain metastases; GFR, glomerular filtration rate; ICU, intensive care unit.
The severity and the outcome may be related both to the multiorgan failure and to the fact that both patients were nephrectomized.
The patient was diagnosed with asymptomatic hypothyroidism during treatment.
The intermittent dosing schedules serve several purposes: (1) Mitigating toxicity: By allowing periods for the body to recover, the intermittent approach helps to reduce the severity and duration of AEs; (2) Maintaining efficacy: Despite the reduced dosing frequency, the therapeutic response could be sustained; and (3) Preventing drug resistance: The strategy may also delay the development of drug resistance by inducing a fitness deficit for drug‐resistant cells during treatment breaks, enhancing the expression of immunostimulating molecules, and reducing immunosuppressive factors, and maintaining the apoptotic and cell cycle arrest effect. 18
High‐intensity exercise is not recommended to patients on TKI treatment because strenuous exercise could promote serum CK elevation and thereby aggravating related symptoms. Patients with workout routines should monitor their serum CK level periodically when receiving high‐dose TKI therapy. They should also be on the watch out for signs and symptoms of rhabdomyolysis, such as unexplained pain, tenderness, weakness, or persistent muscle pain. 41
Factors that influence the pharmacokinetics of TKIs and risk for TKI‐associated CK elevation including high‐dose TKI therapy, polypharmacy and drug–drug interactions. To alleviate high incidence of CK elevation of brigatinib, the phase III first‐line trial introduced brigatinib at a lower dose before dose escalation. 41 Simultaneous use of TKI and statin may induce rhabdomyolysis, especially after long‐period treatment of statin. TKIs inhibiting CYP450 isoenzymes, OATP 1B1, or P‐gp, may affect the metabolism and tissue distribution of statins, and increase their circulating levels and the risk for statin‐associated CK elevation. Treatments can be continued with reduced dose of combination therapy or permanently discontinuation of the suspected statin. It is desirable to assess the benefits of maintaining statin therapy against the potential risk of rhabdomyolysis, cardiovascular events, or other severe symptoms. 22
Mechanism of TKI‐induced serum CK elevation by TKIs in cancer patients
The exact mechanism of TKI‐induced CK elevation is not fully understood and may vary depending on specific TKIs. Several potential mechanisms have been proposed as follows.
Direct myotoxic effect
TKIs are known to elicit a direct toxic effect on muscle cells (myocytes), leading to cell damage and death. This myotoxicity can result in the release of intracellular contents, including CK, into the bloodstream. 95 Platelet‐derived growth factor (PDGF) and its receptor (PDGFR) are involved in the pathophysiology of fiber regeneration, fibrosis, and progression of dystrophy in muscles. For a few specific TKIs, it was reported that PDGFR and c‐abl are inhibited by imatinib mesylate, thereby leading to CK elevation. 15 A significant weight loss has been described in patients taking sorafenib, which was associated with a significant reduction in skeletal muscle mass. Although the mechanism of this muscle loss is unclear, it has been suggested that kinases may play a relevant role in the regulation of muscle protein synthesis. 104 Dasatinib may inhibit the activities of some receptor TKs, including PDGFR. 105
Mitochondrial dysfunction
Some TKIs can interfere with mitochondrial function in muscle cells (Figure 2). 106 For example, osimertinib has the potential to induce mitochondrial dysfunction by influencing the levels of adenosine triphosphate (ATP) and disrupting the activities of the ATP synthase (complex V), leading to a decrease in ATP production. 106 Exposure of cardiomyocyte cultures to sorafenib treatment resulted in an increase in PTX3 expression and triggered cytoskeletal remodeling, diminished contractile capacity, suppression of sodium current, and dysfunction of mitochondrial respiration. 107 Mitochondria are responsible for producing energy in cells, and their dysfunction can lead to cell damage and death, thereby releasing CK.
FIGURE 2.
Underlying mechanisms leading to rhabdomyolysis and serum CK level elevation. (A) Depletion of energy (ATP) which inhibits the function of Na+/K+ ATPase, leading to an increase in intracellular sodium. (B) Increase in intracellular calcium due to the 2Na+/Ca2+ exchanger. (C) Energy depletion also prevents Ca2+ ATPase from pumping out excess calcium, resulting in its accumulation within cells. (D) The buildup of intracellular content of calcium activates proteases (e.g., phospholipase 2 (PLA2)), which break down structural components of cell membranes to facilitate calcium influx. (E) Ultimately, this overload disrupts mitochondrial integrity and triggers apoptosis that leads to muscle cell necrosis. 88
Off‐target kinase inhibition
While TKIs are designed to target specific TKs, they may also inhibit other kinases unintentionally. This off‐target effect can disrupt various cellular processes, including those in muscle cells, leading to increased serum CK levels. 108
Electrolyte disturbances
TKIs can cause imbalances in serum levels of electrolytes (such as potassium, calcium, and phosphate), 109 which are essential for muscle function. Electrolyte imbalances can lead to muscle weakness or damage, which can increase CK levels.
Immune‐mediated mechanisms
The TKI or the damaged muscle tissue may interact with the body immune system to give rise to an inflammatory response, which could further damage muscle cells and elevate CK levels. 110 Afatinib, sorafenib, and ponatinib activate endoplasmic reticulum stress, which leads to cardiotoxicity by promoting the expression of pro‐inflammatory factors and cardiac fetal genes, such as Nfkb1, Il‐6, Tnf, Txnip, and Il1b, through the coordinated activation of the PERK and IRE1α signaling pathways. 111
Ischemia
By affecting blood vessels or blood flow, some TKIs could reduce blood supply (ischemia) to muscles, 112 thereby causing muscle cell damage due to lack of oxygen and nutrients and subsequent CK release. For example, ponatinib and nilotinib have been associated with an increased risk of myocardial ischemia or infarction. 113 Furthermore, crizotinib has been shown to impair the autophagy process, leading to cardiomyocyte death and cardiac injury through the inhibition of MET protein degradation. Reduced autophagy activity has been observed in various cardiac diseases, including ischemia–reperfusion injury, myocardial infarction, cardiac hypertrophy, and heart failure. 114
Drug–drug interactions
The concomitant consumption of TKIs and other drugs could potentiate muscle damage, thereby leading to higher serum CK levels. Several TKIs (e.g., palbociclib) are known to inhibit CYP450 isoenzymes and drug transporters (including OATP 1B1 and P‐gp) (Figure 3), thus increasing the plasma level of statins and the risk for statin‐associated CK elevation. Indeed, the combination of palbociclib and simvastatin has been reported to induce severe rhabdomyolysis. 26 Palbociclib is a weak CYP3A4 inhibitor. In clinical drug–drug interaction studies, palbociclib at 125 mg daily increased the mean C max of midazolam (CYP3A4 substrate) by 37% and AUC values by 61% compared with midazolam alone. 115 Similarly, pharmacokinetics studies have shown that co‐administration of midazolam with multiple doses of ribociclib increases the midazolam plasma concentration by 280%, compared with the administration of midazolam alone. 116 Therefore, the addition of palbociclib/ribociclib may have significantly increased atorvastatin/simvastatin plasma concentration via CYP3A4‐mediated inhibition resulting in statin‐induced rhabdomyolysis. 22
FIGURE 3.
Schematic diagram showing the effect of TKIs (e.g., palbociclib and ribociclib) on uptake of statins into hepatocytes. The uptake of statin (simvastatin) into liver cells is regulated by the organic‐anion‐transporting polypeptide (OATP) transporter. Inside the hepatocytes, statin is metabolized by the liver enzyme CYP3A4. The biotransformed statin is then pumped out of the cells by the ATP‐dependent membrane efflux transporters (ABCB1 and ABCG2). Around 85% of statin is cleared from the body through hepatic excretion via hepatocytes and bile, while approximately 10% is eliminated through renal clearance. The remaining 5% represents the bioavailable and active form of the drug in blood circulation. A specific genetic variant rs4149056 in the SLCO1B1 gene is known to remarkably reduce the transport activity of the OATP transporter. Therefore, patients harboring the SLCO1B1 SNP rs4149056 are expected to exhibit reduced hepatic uptake of statin and thus leading to an increased statin bioavailability. On the contrary, TKIs could also inhibit the CYP3A4 enzyme to reduce the extent of statin metabolism. 26
Genetic predisposition
Certain genetic polymorphisms may predispose some patients TKI‐mediated muscle damage. The single nucleotide polymorphism (rs4149056) of the hepatic uptake transporter OATP1B1 was associated with a higher chance of progressive rhabdomyolysis in patients receiving palbociclib and simvastatin simultaneously. 26 These mechanisms are not mutually exclusive, and a combination of them may be responsible for the increase in serum CK levels in patients taking TKIs. Further research is necessary to understand the full spectrum of effects that TKIs have on muscle tissue and the mechanisms leading to serum CK elevation.
CONCLUSION AND FUTURE PERSPECTIVES
The elevation of serum CK levels in cancer patients on TKI therapy is a notable clinical manifestation that could hinder the anticancer efficacy. This review provided a latest update about potential mechanisms underlying TKI‐induced CK elevation, its clinical implications, and the utility of monitoring serum CK levels for predicting adverse effects.
The physiological function of CK is vital for cellular energy metabolism, muscle contraction, and ATP regeneration. CK is distributed widely throughout the human body, with different subtypes found in various tissues. The elevation of serum CK levels in individual subject can be attributed to multiple factors, including age, gender, race, geographical location, and exercise. Pathological conditions affecting muscle tissues, such as neuromuscular disorders and cardiovascular diseases, and certain medications, including statins, antipsychotics, and antivirals, can also contribute to elevated CK levels.
Molecular targeted TKIs have been found to induce CK elevation in over 20% of cancer patients. It is noteworthy that CK elevation is not preferentially induced by any specific classes of TKIs. The clinical implications of TKI‐induced CK elevation are significant. Although under certain circumstance, mostly asymptomatic, elevated CK levels may also manifest as more severe symptoms such as myopathy, myositis, cardiotoxicity, rhabdomyolysis, rash, and acneiform dermatitis. The occurrence of these symptoms may necessitate dose reduction of TKI treatment and therefore hindering antitumor efficacy.
The underlying mechanisms driving TKI‐induced serum CK elevation are not fully understood. Numerous factors contribute to this phenomenon. CK is an essential enzyme involved in cellular energy metabolism and muscle function. TKIs may disrupt the delicate balance of cellular processes, leading to muscle damage and subsequent release of cellular CK into the bloodstream. TKIs may also affect mitochondrial function and alter ATP/ADP transport, further impacting CK levels. Other factors, including pre‐existing risk factors, high‐dose TKI therapy, polypharmacy, drug–drug interactions (statins), and pharmacogenetic variations, can also influence the pharmacokinetics of TKIs and increase the risk for TKI‐associated CK elevation. Screening for the common rs4149056 polymorphism in SLCO1B1 (a hepatic uptake transporter) could also be a reasonable approach when considering statins and TKIs combination treatments. 26
In the future, several areas warrant attention regarding TKI‐induced CK elevation. Further research is needed to elucidate the precise mechanisms underlying CK elevation in response to specific TKIs. Understanding these mechanisms will aid in the development of specific interventions to mitigate CK‐related adverse effects. Identifying predictive markers or genetic variants associated with TKI‐induced CK elevation may help personalize treatment strategies and optimize patient care. Moreover, novel therapeutic agents could be developed to mitigate muscle damage and preserve CK homeostasis. The clinical management of TKI‐induced CK elevation is currently based on symptomatic management and close monitoring of serum CK levels. Enhancing patient awareness regarding TKI‐induced CK elevation is vital. Regular monitoring of serum CK levels, along with clinical assessment of other potential adverse effects, can help guide treatment decisions and ensure patient safety. The presence of significant CK elevation or severe symptoms may necessitate dose adjustment, drug discontinuation, or implementation of additional interventions. Careful dose selection is of paramount importance for patients with elevated CK levels. The principles of quantitative pharmacology can be applied to ensure target inhibition while minimizing off‐target inhibition in this patient population. Pharmacokinetic (PK) and pharmacodynamic (PD) studies can be used to establish the dose–response relationship and identify the optimal dose range that provides the necessary target inhibition. To this end, Gupta et al. 68 recently conducted efficacy and safety exposure‐response analyses of pivotal ALTA studies to support a favorable benefit–risk profile with the approved dosing regimen (180 mg q.d. with 7‐day lead‐in at 90 mg) versus 90 mg q.d. of brigatinib in ALK–Positive NSCLC.
It is noteworthy that the correlation between TKI‐induced serum CK elevation and the antitumor response or prognosis remains to be determined. Multiple studies investigated the relationship between elevated serum CK levels and the effectiveness of TKI therapy in different cancer types. Interestingly, CK elevation is often associated with improved overall survival (OS) and event‐free survival (EFS) in CML patients receiving TKIs (such as imatinib, dasatinib, nilotinib, and bosutinib). 52 In NSCLC patients with EGFR mutations, serum CK elevation following aumolertinib is also linked to longer PFS and better treatment response. 110 , 117 , 118 , 119 Collectively, higher baseline serum CK levels and significant CK increase after TKI treatment were found to be positive prognostic indicators. 110 These suggested that the extent of CK elevation may have predictive value in both adverse effects and efficacy. In clinical setting, close monitoring of serum CK level may allow physicians to strike a balance between efficacy and adverse effect before dose adjustment or treatment discontinuation.
In summary, TKI‐induced CK elevation is a prevalent adverse effect in cancer patients on targeted therapy. Serum CK level may serve as a valuable clinical biomarker for monitoring treatment response and detecting potential adverse effects. Future research should focus on unraveling the underlying mechanisms, exploring the potential of CK monitoring as a prognostic biomarker, and developing strategies to preserve CK homeostasis. Improved understanding of TKI‐induced CK elevation will contribute to the development of personalized treatment strategies and enhance patient care in the era of targeted anticancer therapies.
FUNDING INFORMATION
No funding was received for this work.
CONFLICT OF INTEREST STATEMENT
The authors declared no competing interests for this work.
Zhang H, To KKW. Serum creatine kinase elevation following tyrosine kinase inhibitor treatment in cancer patients: Symptoms, mechanism, and clinical management. Clin Transl Sci. 2024;17:e70053. doi: 10.1111/cts.70053
REFERENCES
- 1. Moghadam‐Kia S, Oddis CV, Aggarwal R. Approach to asymptomatic creatine kinase elevation. Cleve Clin J Med. 2016;83:37‐42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Ramalingam SS, Vansteenkiste J, Planchard D, et al. Overall survival with osimertinib in untreated, EGFR‐mutated advanced NSCLC. N Engl J Med. 2020;382(1):41‐50. [DOI] [PubMed] [Google Scholar]
- 3. Wallimann T, Tokarska‐Schlattner M, Schlattner U. The creatine kinase system and pleiotropic effects of creatine. Amino Acids. 2011;40(5):1271‐1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Monk BJ, Grisham RN, Banerjee S, et al. MILO/ENGOT‐ov11: Binimetinib versus physician's choice chemotherapy in recurrent or persistent low‐grade serous carcinomas of the ovary, fallopian tube, or primary peritoneum. J Clin Oncol. 2020;38(32):3753‐3762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Yang JC, Liu G, Lu S, et al. Brigatinib versus alectinib in ALK‐positive NSCLC after disease progression on crizotinib: results of phase 3 ALTA‐3 trial. J Thorac Oncol. 2023;18(12):1743‐1755. [DOI] [PubMed] [Google Scholar]
- 6. Ascierto PA, McArthur GA, Dréno B, et al. Cobimetinib combined with vemurafenib in advanced BRAF(V600)‐mutant melanoma (coBRIM): updated efficacy results from a randomised, double‐blind, phase 3 trial. Lancet Oncol. 2016;17(9):1248‐1260. [DOI] [PubMed] [Google Scholar]
- 7. Sugimoto H, Matsumoto S, Tsuji Y, Sugimoto K. Elevated serum creatine kinase levels due to osimertinib: a case report and review of the literature. J Oncol Pharm Pract. 2022;28(2):489‐494. [DOI] [PubMed] [Google Scholar]
- 8. Sakamoto T, Saito Y, Takekuma Y, Kikuchi E, Sugawara M. Gefitinib‐induced myositis: a novel case report. Yakugaku Zasshi. 2023;143(7):617‐620. [DOI] [PubMed] [Google Scholar]
- 9. Fujioka S, Kitajima T, Itotani R. Myositis in a patient with advanced lung cancer treated with osimertinib. J Thorac Oncol. 2018;13(8):e137‐e139. [DOI] [PubMed] [Google Scholar]
- 10. Parafianowicz P, Krishan R, Beutler BD, Islam RX, Singh T. Myositis ‐ a common but underreported adverse effect of osimertinib: case series and review of the literature. Cancer Treat Res Commun. 2020;25:100254. [DOI] [PubMed] [Google Scholar]
- 11. AlJohani NI, Carette S, Lipton JH. Inclusion body myositis in a patient with chronic myeloid leukemia treated with dasatinib: a case report. J Med Case Rep. 2015;9:214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Spechbach H, Morel P, Ing Lorenzini K, et al. Reversible ventricular arrythmia induced by dasatinib. Clin Case Rep. 2013;1(1):20‐25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Guo CW, Alexander M, Dib Y, et al. A closer look at immune‐mediated myocarditis in the era of combined checkpoint blockade and targeted therapies. Eur J Cancer. 2020;124:15‐24. [DOI] [PubMed] [Google Scholar]
- 14. Moscetti L, Nelli F, Ruggeri EM. Rhabdomyolysis from erlotinib: a case report. Tumori. 2011;97(3):415‐416. [DOI] [PubMed] [Google Scholar]
- 15. Uz B, Dolasik I. An unexpected and devastating adverse event of dasatinib: rhabdomyolysis. Leuk Res Rep. 2015;5:1‐2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Gordon JK, Magid SK, Maki RG, Fleisher M, Berman E. Elevations of creatine kinase in patients treated with imatinib mesylate (Gleevec). Leuk Res. 2010;34(6):827‐829. [DOI] [PubMed] [Google Scholar]
- 17. Penel N, Blay JY, Adenis A. Imatinib as a possible cause of severe rhabdomyolysis. N Engl J Med. 2008;358(25):2746‐2747. [DOI] [PubMed] [Google Scholar]
- 18. Matter AV, Micaletto S, Urner‐Bloch U, Dummer R, Goldinger SM. Long‐term response to intermittent binimetinib in patients with NRAS‐mutant melanoma. Oncologist. 2020;25(11):e1593‐e1597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Gettinger SN, Huber RM, Kim DW, et al. Long‐term efficacy and safety of brigatinib in crizotinib‐refractory ALK+ NSCLC: final results of the phase 1/2 and randomized phase 2 (ALTA) trials. JTO Clin Res Rep. 2022;3(9):100385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Logue JM, Kiani B, Bitting RL. Pazopanib and statin‐induced rhabdomyolysis. Case Rep Oncol. 2017;10(3):954‐957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Badran O, Abu Amna M, Turgeman I, Bar‐Sela G. Rhabdomyolysis induced by the interaction between ribociclib and statins‐ case report and literature review. Breast Cancer. 2023;15:47‐50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Nelson KL, Stenehjem D, Driscoll M, Gilcrease GW. Fatal statin‐induced rhabdomyolysis by possible interaction with palbociclib. Front Oncol. 2017;7:150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Beyer IW, Karmali R, Demeester‐Mirkine N, et al. Serum creatine kinase levels in overt and subclinical hypothyroidism. Thyroid. 1998;8:1029‐1031. [DOI] [PubMed] [Google Scholar]
- 24. Rodolico C, Toscano A, Benvenga S, et al. Myopathy as the persistently isolated symptomatology of primary autoimmune hypothyroidism. Thyroid. 1998;8:1033‐1038. [DOI] [PubMed] [Google Scholar]
- 25. Scott KR, Simmons Z, Boyer PJ. Hypothyroid myopathy with strikingly elevated serum creatine kinase level. Muscle Nerve. 2002;26:141‐144. [DOI] [PubMed] [Google Scholar]
- 26. Nersesjan V, Hansen K, Krag T, Duno M, Jeppesen TD. Palbociclib in combination with simvastatin induce severe rhabdomyolysis: a case report. BMC Neurol. 2019;19(1):247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Ide T, Brown‐Endres L, Chu K, et al. GAMT, a p53‐inducible modulator of apoptosis, is critical for the adaptive response to nutrient stress. Mol Cell. 2009;36(3):379‐392. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 28. Stadhouders AM, Jap PH, Winkler HP, Eppenberger HM, Wallimann T. Mitochondrial creatine kinase: a major constituent of pathological inclusions seen in mitochondrial myopathies. Proc Natl Acad Sci USA. 1994;91(11):5089‐5093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Zhong RH, Chen XL, Zeng H, et al. The value of creatine kinase/creatine kinase isoenzyme in differentiating myocardial injury from non‐myocardial injury. Chin J Hygiene Lab. 2017;22:101‐110. [Google Scholar]
- 30. Dongping C. The mechanism of skeletal muscle cell injury in sports. J Xi'an Univ Arts Sci. 2007;04:16‐19. [Google Scholar]
- 31. Hua P, Xuehong Y, Yongjun W. The relationship between serum creatine kinase levels and clinical symptoms in patients with hypokalemic periodic paralysis. J Cap Med Univ. 2003;24(2):178‐179. [Google Scholar]
- 32. Morandi L, Angelini C, Prelle A, et al. High plasma creatine kinase: review of the literature and proposal for a diagnostic algorithm. Neurol Sci. 2006;27(5):303‐311. [DOI] [PubMed] [Google Scholar]
- 33. Brancaccio P, Maffulli N, Limongelli FM. Creatine kinase monitoring in sport medicine. Br Med Bull. 2007;81‐82:209‐230. [DOI] [PubMed] [Google Scholar]
- 34. Venance SL. Approach to the patient with hyperCKemia. Continuum (Minneap Minn). 2016;22(6):1803‐1814. [DOI] [PubMed] [Google Scholar]
- 35. Bruera E. ABC of palliative care. Anorexia, cachexia, and nutrition. BMJ. 1997;315(7117):1219‐1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Iwata Y, Suzuki N, Ohtake H, et al. Cancer cachexia causes skeletal muscle damage via transient receptor potential vanilloid 2‐independent mechanisms, unlike muscular dystrophy. J Cachexia Sarcopenia Muscle. 2016;7(3):366‐376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Tomaszewski M, Stępień KM, Tomaszewska J, Czuczwar SJ. Statin‐induced myopathies. Pharmacol Rep. 2011;63(4):859‐866. [DOI] [PubMed] [Google Scholar]
- 38. Mancini GB, Baker S, Bergeron J, et al. Diagnosis, prevention, and management of statin adverse effects and intolerance: Canadian consensus working group update (2016). Can J Cardiol. 2016;32(7 Suppl):S35‐S65. [DOI] [PubMed] [Google Scholar]
- 39. Laoutidis ZG, Kioulos KT. Antipsychotic‐induced elevation of creatine kinase: a systematic review of the literature and recommendations for the clinical practice. Psychopharmacology. 2014;231(22):4255‐4270. [DOI] [PubMed] [Google Scholar]
- 40. Monteiro P, Perez I, Pich J, Gatell JM, Martínez E. Creatine kinase elevation in HIV‐1‐infected patients receiving raltegravir‐containing antiretroviral therapy: a cohort study. J Antimicrob Chemother. 2013;68(2):404‐408. [DOI] [PubMed] [Google Scholar]
- 41. Camidge DR, Kim HR, Ahn MJ, et al. Brigatinib versus crizotinib in ALK inhibitor‐naive advanced ALK‐positive NSCLC: final results of phase 3 ALTA‐1L trial. J Thorac Oncol. 2021;16(12):2091‐2108. [DOI] [PubMed] [Google Scholar]
- 42. Shi Y, Chen J, Zhang H, et al. Efficacy and safety of iruplinalkib (WX‐0593) in ALK‐positive crizotinib‐resistant advanced non‐small cell lung cancer patients: a single‐arm, multicenter phase II study (INTELLECT). BMC Med. 2023;21(1):72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Dummer R, Flaherty KT, Robert C, et al. COLUMBUS 5‐year update: a randomized, open‐label, phase III trial of encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF V600‐mutant melanoma. J Clin Oncol. 2022;40(36):4178‐4188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Tabernero J, Grothey A, Van Cutsem E, et al. Encorafenib plus cetuximab as a new standard of care for previously treated BRAF V600E‐mutant metastatic colorectal cancer: updated survival results and subgroup analyses from the BEACON study. J Clin Oncol. 2021;39(4):273‐284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Ascierto PA, Schadendorf D, Berking C, et al. MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non‐randomised, open‐label phase 2 study. Lancet Oncol. 2013;14(3):249‐256. [DOI] [PubMed] [Google Scholar]
- 46. Watanabe K, Otsu S, Hirashima Y, et al. A phase I study of binimetinib (MEK162) in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol. 2016;77(6):1157‐1164. [DOI] [PubMed] [Google Scholar]
- 47. Bardia A, Gounder M, Rodon J, et al. Phase Ib study of combination therapy with MEK inhibitor binimetinib and phosphatidylinositol 3‐kinase inhibitor buparlisib in patients with advanced solid tumors with RAS/RAF alterations. Oncologist. 2020;25(1):e160‐e169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Gross AM, Dombi E, Wolters PL, et al. Long‐term safety and efficacy of selumetinib in children with neurofibromatosis type 1 on a phase 1/2 trial for inoperable plexiform neurofibromas. Neuro‐Oncology. 2023;25(10):1883‐1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Bedard PL, Tabernero J, Janku F, et al. A phase Ib dose‐escalation study of the oral pan‐PI3K inhibitor buparlisib (BKM120) in combination with the oral MEK1/2 inhibitor trametinib (GSK1120212) in patients with selected advanced solid tumors. Clin Cancer Res. 2015;21(4):730‐738. [DOI] [PubMed] [Google Scholar]
- 50. Adenis A, Bouché O, Bertucci F, et al. Serum creatine kinase increase in patients treated with tyrosine kinase inhibitors for solid tumors. Med Oncol. 2012;29(4):3003‐3008. [DOI] [PubMed] [Google Scholar]
- 51. Gambacorti‐Passerini C, Cortes JE, Lipton JH, et al. Safety of bosutinib versus imatinib in the phase 3 BELA trial in newly diagnosed chronic phase chronic myeloid leukemia. Am J Hematol. 2014;89(10):947‐953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Bankar A, Lipton JH. Association of creatine kinase elevation with clinical outcomes in chronic myeloid leukemia: a retrospective cohort study. Leuk Lymphoma. 2022;63(1):179‐188. [DOI] [PubMed] [Google Scholar]
- 53. Franceschino A, Tornaghi L, Benemacher V, Assouline S, Gambacorti‐Passerini C. Alterations in creatine kinase, phosphate and lipid values in patients with chronic myeloid leukemia during treatment with imatinib. Haematologica. 2008;93(2):317‐318. [DOI] [PubMed] [Google Scholar]
- 54. Réa D, Mauro MJ, Boquimpani C, et al. A phase 3, open‐label, randomized study of asciminib, a STAMP inhibitor, vs bosutinib in CML after 2 or more prior TKIs. Blood. 2021;138(21):2031‐2041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3‐mutated AML. N Engl J Med. 2019;381(18):1728‐1740. [DOI] [PubMed] [Google Scholar]
- 56. Usuki K, Sakura T, Kobayashi Y, et al. Clinical profile of gilteritinib in Japanese patients with relapsed/refractory acute myeloid leukemia: an open‐label phase 1 study. Cancer Sci. 2018;109(10):3235‐3244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Lu S, Dong X, Jian H, et al. AENEAS: a randomized phase III trial of aumolertinib versus gefitinib as first‐line therapy for locally advanced or metastatic non‐small‐cell lung cancer with EGFR exon 19 deletion or L858R mutations. J Clin Oncol. 2022;40(27):3162‐3171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Malik PS, Noronha V, Dabkara D, et al. Safety of osimertinib in adult patients with metastatic epidermal growth factor receptor T790M mutation‐positive non‐small cell lung cancer: results from a phase IV study in India. Indian J Cancer. 2022;59(Supplement):S1‐S10. [DOI] [PubMed] [Google Scholar]
- 59. Xiong A, Ren S, Liu H, et al. Efficacy and safety of SH‐1028 in patients with EGFR T790M‐positive NSCLC: a multicenter, single‐arm, open‐label, phase 2 trial. J Thorac Oncol. 2022;17(10):1216‐1226. [DOI] [PubMed] [Google Scholar]
- 60. Wang M, Fan Y, Sun M, et al. Sunvozertinib for the treatment of NSCLC with EGFR Exon20 insertion mutations: the first pivotal study results. J Clin Oncol. 2023;41(suppl 16):9002. [Google Scholar]
- 61. Motzer RJ, Hutson TE, Tomczak P, et al. Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J Clin Oncol. 2009;27(22):3584‐3590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Yamanaka T, Takemura K, Hayashida M, Suyama K, Urakami S, Miura Y. Cabozantinib‐induced serum creatine kinase elevation and rhabdomyolysis: a retrospective case series. Cancer Chemother Pharmacol. 2023;92(3):235‐240. [DOI] [PubMed] [Google Scholar]
- 63. Okubo H, Ando H, Takasaki Y, et al. Impact of cabozantinib exposure on proteinuria and muscle toxicity in patients with unresectable hepatocellular carcinoma. Pharmaceuticals (Basel). 2022;15(12):1460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Goyal L, Meric‐Bernstam F, Hollebecque A, et al. Futibatinib for FGFR2‐rearranged intrahepatic cholangiocarcinoma. N Engl J Med. 2023;388(3):228‐239. [DOI] [PubMed] [Google Scholar]
- 65. Li J, Zhang X, Deng Y, et al. Efficacy and safety of avapritinib in treating unresectable or metastatic gastrointestinal stromal tumors: a phase I/II, open‐label, multicenter study. Oncologist. 2023;28(2):187 ‐e114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Saleh M, Cassier PA, Eberst L, et al. Phase I study of ramucirumab plus merestinib in previously treated metastatic colorectal cancer: safety, preliminary efficacy, and pharmacokinetic findings. Oncologist. 2020;25(11):e1628‐e1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Schoffski P, Cho BC, Italiano A, et al. BOS172738, a highly potent and selective RET inhibitor, for the treatment of RET‐altered tumors including RET‐fusion+ NSCLC and RET‐mutant MTC: phase 1 study results. J Clin Oncol. 2021;39(15 suppl):3008. [Google Scholar]
- 68. Gupta N, Wang X, Offman E, et al. Brigatinib dose rationale in anaplastic lymphoma kinase‐positive non‐small cell lung cancer: exposure‐response analyses of pivotal ALTA study. CPT Pharmacometrics Syst Pharmacol. 2020;9(12):718‐730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Gupta N, Hanley MJ, Kerstein D, et al. Effect of several renal impairment on the pharmacokinetics of brigatinib. Investig New Drugs. 2021;39:1306‐1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Huang WS, Liu S, Zou D, et al. Discovery of brigatinib (AP26113), a phosphine oxide‐containing, potent, orally active inhibitor of anaplastic lymphoma kinase. J Med Chem. 2016;59:4948‐4964. [DOI] [PubMed] [Google Scholar]
- 71. Morcos PN, Nueesch E, Jaminion F, et al. Exposure‐response analysis of alectinib in crizotinib‐resistant ALK‐positive non‐small cell lung cancer. Cancer Chemother Pharmacol. 2018;82:129‐138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Sakamoto H, Tsukaguchi T, Hiroshima S, et al. CH5424802, a selective ALK inhibitor capable of blocking the resistant gatekeeper mutant. Cancer Cell. 2011;19:679‐690. [DOI] [PubMed] [Google Scholar]
- 73. Li B, Liu S, Feng H, et al. Prediction of trough concentration and ALK occupancy in plasma and cerebrospinal fluid using physiologically based pharmacokinetic modeling of crizotinib, alectinib, and lorlatinib. Front Pharmacol. 2023;14:1234262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Zou HY, Li Q, Lee JH, et al. An orally available small‐molecule inhibitor of c‐met, PF‐2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res. 2007;67:4408‐4417. [DOI] [PubMed] [Google Scholar]
- 75. Remsing Rix LL, Rix U, Colinge J, et al. Global target profile of the kinase inhibitor bosutinib in primary chronic myeloid leukemia cells. Leukemia. 2009;23(3):477‐485. [DOI] [PubMed] [Google Scholar]
- 76. Zhang X, Zhang M, Du X, et al. Clinical efficacy and safety analysis of aumolertinib in real‐world treatment of EGFR‐mutated advanced non‐small‐cell lung cancer. Front Pharmacol. 2024;15:1331138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Barnes TA, O'Kane GM, Vincent MD, Leighl NB. Third generation tyrosine kinase inhibitors targeting epidermal growth factor receptor mutations in non‐small cell lung cancer. Front Oncol. 2017;7:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Yang JC, Ahn MJ, Kim DW. Osimertinib in pretreated T790M‐positive advanced non–small‐cell lung cancer: AURA study phase II extension component. J Clin Oncol. 2017;35(12):1288‐1296. [DOI] [PubMed] [Google Scholar]
- 79. Akamatsu H, Katakami N, Okamoto I, et al. Osimertinib in Japanese patients with EGFR T790M mutation positive advanced non‐small‐cell lung cancer: AURA3 trial. Cancer Sci. 2018;109(6):1930‐1938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in untreated EGFR‐mutated advanced non‐small‐cell lung cancer. N Engl J Med. 2018;378(2):113‐125. [DOI] [PubMed] [Google Scholar]
- 81. Harati Y, Biliciler S. Myopathies. In: Rolak KA, ed. Neurology Secrets. 5th ed. Mosby: Elsevier; 2010:63‐82. [Google Scholar]
- 82. Crowley F, Fitzgerald BG, Bhardwaj AS, Siraj I, Smith C. Life‐threatening myositis in a patient with EGFR‐mutated NSCLC on osimertinib: case report. JTO Clin Res Rep. 2021;3(1):100260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Kwok WC, Tam TC, Lui MM, Lam DC, Ho JC. Control of brain metastases with alectinib in anaplastic lymphoma kinase‐rearranged lung cancer. Respirol Case Rep. 2017;5(3):e00224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Binzaid AA, Baqal OJ, Soheib M, Al Nahedh M, Samarkandi HH, Aljurf M. Cardiovascular toxicity associated with tyrosine kinase inhibitor therapy in chronic myeloid leukemia. Gulf J Oncolog. 2021;1(37):79‐84. [PubMed] [Google Scholar]
- 85. Al‐Ali HK, Krahl R, Orth M, et al. Persistently increased creatine kinase levels in patients with chronic myeloid leukemia treated with imatinib correlate with major cytogenetic remission. Blood. 2004;104(11):2933.15238418 [Google Scholar]
- 86. Chavez LO, Leon M, Einav S, Varon J. Beyond muscle destruction: a systematic review of rhabdomyolysis for clinical practice. Crit Care. 2016;20(1):135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Augustyn K, Joseph J, Patel AB, Razmandi A, Ali AN, Tawbi HA. Treatment experience with encorafenib plus binimetinib for BRAF V600‐mutant metastatic melanoma: management insights for clinical practice. Melanoma Res. 2023;33:406‐416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Streicher C, Daulange A, Madranges N, Vayre L. Severe rhabdomyolysis induced by possible drug‐drug interaction between Ribociclib and simvastatin. J Oncol Pharm Pract. 2021;27(3):722‐726. [DOI] [PubMed] [Google Scholar]
- 89. Moreno Garcia V, Thavasu P, Blanco Codesido M, et al. Association of creatine kinase and skin toxicity in phase I trials of anticancer agents. Br J Cancer. 2012;107:1797‐1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Genentech . Cotellic (cobimetinib). 2015. U.S.Food and Drug Administration. Accessed September 27, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/206192s005lbl.pdf
- 91. Array BioPharma . Mektovi (binimetinib). 2018. U.S.Food and Drug Administration. Accessed September 27, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210498lbl.pdf
- 92. Array BioPharma . Braftov (encorafenib). 2018. U.S.Food and Drug Administration. Accessed September 27, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/210496s014lbl.pdf
- 93. Pharma A. Xospata (gilteritinib). 2018. U.S.Food and Drug Administration. Accessed September 27, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/211349s001lbl.pdf
- 94. Ruggeri EM, Cecere FL, Moscetti L, Doni L, Padalino D, Di Costanzo F. Severe rhabdomyolysis during sunitinib treatment of metastatic renal cell carcinoma. A report of two cases. Ann Oncol. 2010;21(9):1926‐1927. [DOI] [PubMed] [Google Scholar]
- 95. Tsuji K, Takemura K, Minami K, et al. A case of rhabdomyolysis related to sorafenib treatment for advanced hepatocellular carcinoma. Clin J Gastroenterol. 2013;6(3):255‐257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Li Y, Liu Y, Zhao Z, Zhang Y. Rhabdomyolysis in a patient with advanced lung cancer treated with osimertinib: a case report. Transl Lung Cancer Res. 2023;12(3):629‐636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Samaran Q, Belakebi D, Theret S, et al. Hemophagocytic lymphohistiocytosis in advanced melanoma treated with dabrafenib and trametinib combination: two cases. Melanoma Res. 2020;30(5):519‐523. [DOI] [PubMed] [Google Scholar]
- 98. Muto Y, Ng W, Namikawa K, et al. Success of rechallenging dabrafenib and trametinib combination therapy after trametinib‐induced rhabdomyolysis: a case report. Melanoma Res. 2018;28(2):151‐154. [DOI] [PubMed] [Google Scholar]
- 99. Irimada M, Fujimura T, Kambayashi Y, et al. Severe rhabdomyolysis developing in an advanced melanoma patient treated by pembrolizumab followed by dabrafenib trametinib combined therapy. J Dermatol. 2019;46(7):e256‐e258. [DOI] [PubMed] [Google Scholar]
- 100. Finsterer J. Trametinib and dabrafenib induced rhabdomyolysis, renal failure, and visual loss. Report of one case. Rev Med Chile. 2020;148(11):1684‐1689. [DOI] [PubMed] [Google Scholar]
- 101. Zipser MC, Eichhoff OM, Widmer DS, et al. A proliferative melanoma cell phenotype is responsive to RAF/MEK inhibition independent of BRAF mutation status. Pigment Cell Melanoma Res. 2011;24(2):326‐333. [DOI] [PubMed] [Google Scholar]
- 102. Deken MA, Gadiot J, Jordanova ES, et al. Targeting the MAPK and PI3K pathways in combination with PD1 blockade in melanoma. Onco Targets Ther. 2016;5(12):e1238557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Choi H, Deng J, Li S, et al. Pulsatile MEK inhibition improves anti‐tumor immunity and T cell function in murine Kras mutant lung cancer. Cell Rep. 2019;27(3):806‐819.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Zhao Y, Haginoya K, Sun G, et al. Platelet‐derived growth factor and its receptors are related to the progression of human muscular dystrophy: an immunohistochemical study. J Pathol. 2003;201:149‐159. [DOI] [PubMed] [Google Scholar]
- 105. Chen Z, Lee FY, Bhalla KN, et al. Potent inhibition of platelet‐derived growth factor‐induced responses in vascular smooth muscle cells by BMS‐354825 (dasatinib). Mol Pharmacol. 2006;69:1527‐1533. [DOI] [PubMed] [Google Scholar]
- 106. Yang H, Qiu S, Yao T, et al. Transcriptomics coupled with proteomics reveals osimertinib‐induced myocardial mitochondrial dysfunction. Toxicol Lett. 2024;397:23‐33. [DOI] [PubMed] [Google Scholar]
- 107. Chen YT, Masbuchin AN, Fang YH, et al. Pentraxin 3 regulates tyrosine kinase inhibitor‐associated cardiomyocyte contraction and mitochondrial dysfunction via ERK/JNK signalling pathways. Biomed Pharmacother. 2023;157:113962. [DOI] [PubMed] [Google Scholar]
- 108. Hasinoff BB, Patel D, Wu X. The myocyte‐damaging effects of the BCR‐ABL1‐targeted tyrosine kinase inhibitors increase with potency and decrease with specificity. Cardiovasc Toxicol. 2017;17(3):297‐306. [DOI] [PubMed] [Google Scholar]
- 109. Muhammad DP, Ariyah YA, et al. The effect of imatinib and nilotinib on blood calcium and blood potassium levels in chronic myeloid leukemia patient: a literature review. Oncol Rev. 2021;15(2):547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Jiang Y, Su Z, Lin Y, et al. Prognostic and predictive impact of creatine kinase level in non‐small cell lung cancer treated with tyrosine kinase inhibitors. Transl Lung Cancer Res. 2021;10(9):3771‐3781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Wang H, Wang Y, Li J, et al. Three tyrosine kinase inhibitors cause cardiotoxicity by inducing endoplasmic reticulum stress and inflammation in cardiomyocytes. BMC Med. 2023;21(1):147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Escudier B, Eisen T, Stadler WM, et al. Sorafenib in advanced clear‐cell renal‐cell carcinoma. N Engl J Med. 2007;356(2):125‐134. [DOI] [PubMed] [Google Scholar]
- 113. Shyam Sunder S, Sharma UC, Pokharel S. Adverse effects of tyrosine kinase inhibitors in cancer therapy: pathophysiology, mechanisms and clinical management. Signal Transduct Target Ther. 2023;8(1):262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Xu Z, Pan Z, Jin Y, et al. Inhibition of PRKAA/AMPK (Ser485/491) phosphorylation by crizotinib induces cardiotoxicity via perturbing autophagosome‐lysosome fusion. Autophagy. 2024;20(2):416‐436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. U.S. National Library of Medicine . IBRANCE – palbociclib capsule. 2016. Kirkland, Quebec: Pfizer Canada Inc. Accessed September 27, 2024. https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=e0e6412f‐50b4‐4fd4‐9364‐62818d121a07
- 116. KISQALI® (ribociclib) . Highlights of prescribing information. Accessed September 27, 2024. https://www.accessdata.fda.gov/drugsatfda.docs/label/2019/209092s004lbl.pdf
- 117. Lu S, Li C, Jian H, et al. Elevated creatine phosphokinase (CPK) as a strong predictor of aumolertinib (Au) treatment response in patients (pts) with advanced non‐small cell lung cancer (NSCLC): post‐hoc analysis of AENEAS. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14–19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023; 83(7_Suppl): Abstract nr 5462.
- 118. Fan Y, Zhu J, He J, et al. 367P high‐dose aumolertinib in EGFR‐mutant NSCLC patients with brain metastases: primary data from ACHIEVE. Ann Oncol. 2022;33(Supp 9):S1585. [Google Scholar]
- 119. Fang W, Bu Q, Wang Q, et al. 373P safety and efficacy of aumolertinib treatment in patients with advanced NSCLC harboring uncommon EGFR mutations: cohort 2. Ann Oncol. 2022;33(Supp 9):S1587‐S1588. [Google Scholar]