Neurological Complications of Targeted Therapies


Agent

Therapeutic target(s)/mechanism of action

Use(s) in cancer treatment

Common neurologic toxicities (≥10% of patients)

Rare neurologic toxicities (<10%)

Monoclonal antibodies

Bevacizumab

VEGF-A

Metastatic colorectal cancer, nonsquamous NSCLC, platinum-resistant ovarian cancer, advanced cervical cancer, metastatic renal cell carcinoma, recurrent glioblastoma


Ischemic stroke, intracranial hemorrhage, RPLS, optic neuropathy

Ramucirumab

VEGFR2

Metastatic NSCLC, advanced or metastatic gastric or GE junction cancer


Headache, cerebral ischemia, RPLS

Ziv-aflibercept

VGFR-1/2 + IgG1 Fc fusion protein

Metastatic colorectal cancer

Headache

Arterial thrombotic events; RPLS

Tyrosine kinase inhibitors

Lenvatinib mesylate

VEGFR, FGFR, RET, KIT, PDGFRα

Progressive radioactive iodine-refractory differentiated thyroid cancer

Headache

Cerebrovascular ischemic event, TIA, intracranial hemorrhage, seizures, RPLS

Sorafenib tosylate

VEGFR, PDGFR, Raf kinase, KIT, FLT-3

Renal cell carcinoma, hepatocellular carcinoma, radioactive iodine-refractory thyroid cancer


Ischemic stroke

Sunitinib

VEGFR, PDGFR, KIT

Renal cell carcinoma, pancreatic neuroendocrine tumors, imatinib-resistant GIST


Ischemic stroke, reversible cognitive dysfunction with extrapyramidal symptoms, RPLS

Pazopanib

VEGFR, PDGFR, FGFR, Itk, c-Fms

Renal cell carcinoma, soft tissue sarcoma

Headache

Stroke, TIA, intracranial hemorrhage, RPLS

Ponatinib

BCR-ABL, VEGFR, PDGFR, FGFR, ephrin receptor, Src family kinases, c-Kit, RET, TIE2, FLT-3

CML, Philadelphia chromosome-positive ALL

Headache, venous and arterial thrombotic events including stroke, peripheral neuropathy


Regorafenib

VEGFR, PDGFR, FGFR, TIE2, KIT, RET, RAF1, BRAF, mutant BRAF

Advanced GIST, metastatic colorectal cancer


Sensory neuropathy, headache, RPLS, transverse myelopathy

Axitinib

VEGFR

Renal cell carcinoma


Arterial thrombotic events, RPLS

Immunomodulatory agents

Thalidomide

Immune mediating and anti-angiogenic properties

Multiple myeloma

Somnolence, Treatment-emergent peripheral neuropathy (dose-dependent and cumulative)

Reversible altered mental status, worsening of preexisting Parkinson’s disease

Lenalidomide

Second-generation thalidomide analog

Multiple myeloma, mantle cell lymphoma, transfusion-dependent anemia in MDS


Peripheral neuropathy, reversible cognitive/memory deficits and aphasia

Pomalidomide

Second-generation thalidomide analog

Multiple myeloma


Peripheral neuropathy, reversible aphasia


AIDP acute inflammatory demyelinating polyneuropathy, ALL acute lymphoblastic leukemia, ALK anaplastic lymphoma kinase, BCR-ABL breakpoint cluster region-Abelson murine leukemia viral oncogene homolog 1, BTK Bruton’s tyrosine kinase, CDK cyclin-dependent kinase, c-Fms colony-stimulating factor 1 receptor , CLL chronic lymphocytic leukemia, CML chronic myelogenous leukemia, EGFR epidermal growth factor receptor, ER estrogen receptor, FLT-3 FMS-like tyrosine kinase 3, GIST gastrointestinal stromal tumor, HDAC histone deacetylase, HER human epidermal growth factor receptor, JAK Janus kinase, MDS myelodysplastic syndrome, MEK mitogen-activated protein kinase kinase, MPD myeloproliferative disorder, mTOR mammalian target of rapamycin, NHL non-Hodgkin lymphoma, NSCLC non-small cell lung cancer, PARP poly (ADP-ribose) polymerase, PDGFR platelet-derived growth factor receptor, PI3K phosphoinositide-3-kinase, RCC renal cell carcinoma, RET REarranged during Transfection, RPLS reversible posterior leukoencephalopathy syndrome, SEGA subependymal giant cell astrocytoma, SMO smoothened, TIA transient ischemic attack, VEGFR vascular endothelial growth factor receptor




Monoclonal Antibodies



Bevacizumab


Bevacizumab is a recombinant, humanized monoclonal antibody against VEGF-A, and is approved by the United States Food and Drug Administration (U.S. FDA) for use in the treatment of metastatic colorectal cancer, advanced nonsquamous non-small cell lung cancer, platinum-resistant ovarian cancer, advanced cervical cancer, metastatic renal cell carcinoma, and recurrent glioblastoma [7]. Although bevacizumab is generally well tolerated, there are a few rare but serious neurologic complications. Use of bevacizumab is associated with an increased rate of intracranial hemorrhage, with incidence ranging from 0.3 to 0.7% in patients with systemic tumors [8, 9]. In patients with brain metastases or primary brain tumors, the incidence is increased and ranges from 0.8 to 3.3%; this is slightly increased from the baseline rate of spontaneous intracranial hemorrhage in patients with these tumors not treated with anti-angiogenic agents (Fig. 16.1) [8, 10, 11]. Patients receiving bevacizumab are thought to have an increased risk of arterial thromboembolic events, including ischemic stroke (Fig. 16.2). In a meta-analysis of 1745 patients with metastatic cancer, treatment with bevacizumab plus chemotherapy in comparison with chemotherapy alone resulted in an increased risk of an arterial thromboembolic event with a hazard ratio of 2.0 and an absolute rate of 5.5 events per 100 person-years. Of 37 patients with thromboembolic events noted in this study, there were 16 cerebrovascular events (stroke or TIA); rates were higher in patients over 65 years old or with a prior arterial thrombotic event. [12]. A retrospective study of recurrent high-grade glioma patients treated with bevacizumab in clinical trials found the incidence of ischemic stroke to be 1.9%, or 0.38 cases per 100 patient-months [13]. Bevacizumab administration also has been associated with the development of the rare clinical-radiologic Posterior Reversible Encephalopathy Syndrome (PRES), also known as the Reversible Posterior Leukoencephalopathy Syndrome (RPLS). This syndrome has been described in association with a variety of medications, including angiogenesis inhibitors, and is characterized by neurological symptoms including headache, visual disturbance, confusion, and seizures, in conjunction with subcortical/white matter abnormalities, including prominent vasogenic edema, especially in the parietal and occipital lobes, on MRI [14, 15]. These symptoms usually resolve with supportive care. Rarely, optic neuropathy has been described in association with bevacizumab, with an incidence of 1.2% in one study [16]. Of note, all of the patients in that study had also received radiation to the brain for glioblastoma, but at doses felt to be safe to the optic nerves; in comparison, the incidence of severe optic neuropathy in patients receiving radiation but not bevacizumab was 0.2% [16]. Another case series reported three patients with glioblastoma previously treated with radiation therapy who developed optic neuropathy following bevacizumab use [17].

A328796_3_En_16_Fig1_HTML.jpg


Fig. 16.1
Bevacizumab-related intracranial hemorrhage. MRI of the brain in a 72-year-old man with a left parietotemporal glioblastoma on bevacizumab therapy, who was found to have evidence of subacute hemorrhage on MRI. The T1-weighted image on the left shows a hyperintense signal abnormality in the left posterior periventricular area, consistent with blood. In the susceptibility weighted imaging (SWI) sequence on the right, there are hypointense signal changes in the left posterior periventricular region, consistent with blood products


A328796_3_En_16_Fig2_HTML.jpg


Fig. 16.2
Bevacizumab-related ischemic stroke. MRI of the brain in a 71-year-old woman with a left frontal anaplastic astrocytoma on bevacizumab therapy, who presented with an episode of slurred speech. The diffusion weighted imaging (DWI) sequences show hyperintense signal abnormalities in the right basal ganglia, consistent with acute ischemia. (Image courtesy of Ivana Vodopivec, M.D., Ph.D., Brigham and Women’s Hospital.)


Ramucirumab


Ramucirumab is a humanized monoclonal antibody against VEGFR2 and was approved by the FDA in 2014 for the treatment of progressive metastatic non-small cell lung cancer (NSCLC) and for the treatment of advanced or metastatic gastric or gastroesophageal junction cancer. Neurologic complications are rare, and include headache in 9% of patients, arterial thromboembolic events (including cerebrovascular accident, cerebral ischemia, cardiac arrest, myocardial infarction) in 1.7%, and RPLS in <0.1% [18].


Ziv-Aflibercept


Ziv-aflibercept is a recombinant fusion protein comprised of VEGF-binding portions from VEGFR-1 and -2 fused to the Fc portion of the human immunoglobulin IgG1. In August 2012, it received FDA approval for use in combination with 5-fluorouracil, leucovorin and irinotecan (the FOLFIRI regimen) in patients with progressive metastatic colorectal cancer after oxaliplatin therapy. Headache is a common side effect; rare but potentially serious neurologic complications include arterial thrombotic events and RPLS [19].


VEGF Receptor Tyrosine Kinase Inhibitors



Lenvatinib Mesylate


Lenvatinib is an oral inhibitor of VEGFR 1, 2, and 3, as well as an inhibitor of fibroblast growth factor receptors (FGFR) 1, 2, 3, and 4, REarranged during Transfection (RET), KIT, and platelet-derived growth factor receptor alpha (PDGFRα). It was approved by the U.S. FDA in February 2015 for the treatment of locally recurrent or metastatic radioactive iodine-refractory differentiated thyroid cancer. In the Phase III clinical trial, headache was common, experienced in 27.6% of treated patients versus 6.1% of patients receiving placebo. About 3.6% of patients experienced a serious neurologic complication, including cerebrovascular ischemic event, TIA, intracranial hemorrhage, seizures or RPLS [20].


Sorafenib Tosylate


Sorafenib is a multikinase inhibitor active against numerous targets, including VEGFR-2 and -3, PDGFR, Raf kinase, KIT and FMS-like tyrosine kinase 3 (FLT-3), and is used in the treatment of renal cell carcinoma, hepatocellular carcinoma, and radioactive iodine-refractory thyroid cancer. The most concerning potential neurologic complication associated with sorafenib use is stroke; a meta-analysis of clinical trials using sorafenib and sunitinib found a threefold increased risk of arterial thrombotic events, including ischemic stroke [21].


Sunitinib


Sunitinib is a multikinase inhibitor with targets including VEGFR, PDGFR, and KIT, and is approved for use in metastatic renal cell carcinoma, progressive pancreatic neuroendocrine tumors, and imatinib-resistant gastrointestinal stromal tumor. Sunitinib has been associated with increased risk of ischemic stroke, with threefold increased risk of arterial thrombotic events, including ischemic stroke , in a meta-analysis of clinical trials using sorafenib and sunitinib [21]. A reversible syndrome of cognitive dysfunction has been described, with symptoms including confusion, language problems, extrapyramidal symptoms, and gait abnormalities [22]. RPLS also has been reported in association with sunitinib therapy [23].


Pazopanib


Pazopanib is a multi-tyrosine kinase inhibitor of VEGFR, PDGFR, KIT, fibroblast growth factor receptor (FGFR), interleukin-2 receptor inducible T-cell kinase (Itk), and colony-stimulating factor 1 receptor (c-Fms) that is FDA approved for the treatment of advanced renal cell carcinoma and advanced soft tissue sarcoma. Headache has been in reported in 10–23% of patients; serious adverse events are rare, including arterial thrombotic events [stroke and transient ischemic attack (TIA)] and central nervous system (CNS) hemorrhage (subarachnoid hemorrhage, intracranial hemorrhage) [2426]. Cases of RPLS also have been described [2729].


Ponatinib


Ponatinib is an oral multi-target tyrosine kinase inhibitor designed as a pan-BCR-ABL inhibitor but also targeting VEGFR, PDGFR, FGFR, ephrin receptor, Src family kinases, c-Kit, RET, tunica interna endothelial cell kinase 2 (TIE2), and FLT-3 [30]. It received accelerated FDA approval in 2012 for the treatment of patients with chronic myeloid leukemia (CML) or Philadelphia chromosome-positive acute lymphoblastic leukemia, whose disease was resistant or intolerant to prior tyrosine kinase inhibitor therapy [30]. Headache was common in clinical trial participants, and arterial thrombotic events were noted, with cerebrovascular events considered to be at least possibly attributable to the drug in 0.7% of patients in the initial study [31]. Additional data gathered after initial FDA approval showed a high incidence of venous and arterial thrombotic events, some of which were fatal or life-threatening, noted in approximately 24% of patients in the phase II trial and 48% of patients in the phase I trial. This led to temporary suspension of ponatinib marketing and sales by the FDA, a black box warning on the drug, and cancelation of a planned randomized trial of imatinib versus ponatinib [31, 32]. Peripheral neuropathy has been reported in 13% of ponatinib-treated patients (2% grade 3–4) [33].


Regorafenib


Regorafenib is an oral multikinase inhibitor active against VEGFR, PDGFR, FGFR, TIE2, KIT, RET, RAF1, BRAF, and mutant BRAF [34]. It is approved by the U.S. Food and Drug Administration for the treatment of advanced gastrointestinal stromal tumors and previously treated metastatic colorectal cancer. Neurologic side effects are rare; in the colorectal cancer trials, sensory neuropathy was reported in 7% of patients receiving regorafenib (vs. 4% receiving placebo) and headache in 5% of regorafenib patients (vs. 3% with placebo) [34]. There are isolated case reports of RPLS [35] and transverse myelopathy (in a patient previously treated with stereotactic body radiation therapy) [36].


Axitinib


Axitinib is a potent, selective, second-generation inhibitor of VEGFR-1, -2, and -3 [37]. It is FDA approved for the treatment of advanced renal cell carcinoma. Neurologic side effects are rare, but as with other agents of this class, serious adverse effects including arterial thrombotic events and RPLS have been described [38].


Immunomodulatory Anti-angiogenic Agents (Thalidomide and Its Analogs)



Thalidomide


Thalidomide was the first immunomodulatory drug approved for use in multiple myeloma, and has potent immune mediating and antiangiogenic properties [39]. Frequent but mild (grade 1–2) neurologic side effects in the phase II multiple myeloma trial included (at the 200 mg/day dose) somnolence in 34%, tingling or numbness in 12%, incoordination in 16%, tremors in 10%, and headache in 12% [40]. The most prominent neurologic complication associated with thalidomide use is peripheral neuropathy, which has been reported in 23–70% of patients, and appears to be dose-dependent (worse with doses in excess of 200 mg per day) and cumulative with longer duration of treatment. Length-dependent sensory neuropathy is most commonly reported, and autonomic neuropathy is thought to mediate some thalidomide-related side effects including constipation, orthostatic hypotension, bradycardia and sexual dysfunction; motor symptoms are rare [41]. Other than somnolence, CNS toxicity is uncommon. There is a reported case of thalidomide-induced altered mental status progressing to coma, promptly reversed with drug discontinuation [42]. Thalidomide has also been associated with worsening of preexisting Parkinson’s disease [43].


Lenalidomide


Lenalidomide is a second-generation immunomodulatory agent derived from thalidomide, initially approved for use in transfusion-dependent anemia in patients with myelodysplastic syndrome, and subsequently approved for use in multiple myeloma and mantle cell lymphoma. In addition to its immunomodulatory effects, it has direct cytotoxic and antiangiogenic properties [39]. In contrast to thalidomide, lenalidomide is much less neurotoxic; although mild to moderate neuropathy during treatment has been reported, the phase III trial of lenalidomide plus dexamethasone in comparison with lenalidomide alone in relapsed multiple myeloma showed no significant increase in the incidence of peripheral neuropathy in lenalidomide-treated patients [41]. Lenalidomide use rarely has been associated with CNS toxicity, with case reports of reversible cognitive decline, memory problems and expressive language difficulties [42].


Pomalidomide


Pomalidomide is a second-generation thalidomide analog approved for the treatment of progressive multiple myeloma. From the neurologic standpoint, it is generally well tolerated, with 9% of patients experiencing grades 1–2 peripheral neuropathy and no reported cases of grades 3–4 neuropathy in clinical trials [44, 45]. CNS toxicity is uncommon, with one case report of reversible expressive language difficulties associated with pomalidomide use [42].



Small Molecule Inhibitors that Do not Target VEGFR


The growing shift from cytotoxic chemotherapy toward targeted molecular therapy has led to a revolution in drug development, with the rise of many new small molecule drugs designed to inhibit specific cancer targets. This was particularly spurred by the discovery of the BCR-ABL translocation [breakpoint cluster region (BCR)—Abelson murine leukemia viral oncogene homolog 1 (ABL)] as a driver of chronic myeloid leukemia, and the successful use of imatinib, a BCR-ABL inhibitor, to treat these patients [46]. Since then, small molecules have been developed to inhibit varied targets that play a role in oncogenesis in different cancers, many of which are kinase inhibitors. Overall, small molecule inhibitors have relatively few neurologic complications as a class. This section will review the neurologic complications that have been described with treatment using these small molecules, including tyrosine kinase inhibitors, serine/threonine kinase inhibitors, lipid kinase inhibitors, histone deacetylase inhibitors, poly (ADP-ribose) polymerase (PARP) inhibitors , smoothened (SMO) inhibitors, and proteasome inhibitors. This information is summarized in Table 16.2.


Table 16.2
Neurological complications of small molecule inhibitors






































































































































































































































Agent

Therapeutic target(s)/mechanism of action

Use(s) in cancer treatment

Common neurologic toxicities (≥10% of patients)

Rare neurologic toxicities (<10%)

Proteasome inhibitors

Bortezomib

Reversible inhibitor of 26S proteasome

Multiple myeloma; mantle cell lymphoma

Peripheral neuropathy (sensory > autonomic > motor)

RPLS

Carfilzomib

Second-generation proteasome inhibitor

Progressive multiple myeloma

Peripheral neuropathy


Tyrosine kinase inhibitors

Erlotinib Hydrochloride

EGFR

NSCLC

None

Stroke (in combination with gemcitabine)

Gefitinib

EGFR

NSCLC

None


Afatinib Dimaleate

EGFR, HER2, HER4

NSCLC

None


Lapatinib Ditosylate

EGFR1, HER2

HER2 overexpressing breast cancer

Headache


Vandetanib

EGFR, RET, VEGFR2

Medullary thyroid cancer

Headache

Back pain, RPLS, cerebral ischemia, TIA

Crizotinib

ALK, MET, ROS1

ALK-rearranged NSCLC

Visual changes, dizziness


Ceritinib

ALK

ALK-rearranged NSCLC

Headache, dizziness

Muscle spasms, dysphonia, tremor, convulsion

Cabozantinib

MET, VEGFR2, RET

Medullary thyroid cancer

Dysphonia, back pain, headache, dizziness

RPLS, hemorrhage, venous and arterial thrombosis

Imatinib

BCR-ABL, PDGFR, c-Fms, c-Kit

CML, ALL, chronic eosinophilic leukemia, dermatofibrosarcoma protuberans, GIST, MDS/MPD, systemic mastocytosis

Headache, muscle spasms

Intraparenchymal hemorrhage, subdural hemorrhage

Nilotinib

BCR-ABL

CML

Headache

Muscle spasm, CNS hemorrhage

Dasatinib

BCR-ABL, SRC family kinases (Lyn, Src)

ALL, CML

Headache

Subdural hemorrhage

Bosutinib

SRC/ABL1

CML

Headache

Muscle spasms

Ponatinib

BCR-ABL, VEGFR, PDGFR, FGFR, ephrin receptor, Src family kinases, c-Kit, RET, TIE2, FLT-3

CML, Philadelphia chromosome-positive ALL

Headache, venous and arterial thrombotic events including stroke, peripheral neuropathy


Ibrutinib

BTK

CLL, mantle cell lymphoma, Waldenström’s macroglobulinemia

Back pain

Subdural hematoma, confusional state

Ruxolitinib

JAK1 and 2

Polycythemia vera, myelofibrosis

Headache, muscle spasms, dizziness


Serine/threonine kinase inhibitors

Vemurafenib

BRAF

Melanoma

Headache

Facial palsy, AIDP

Dabrafenib

BRAF

Melanoma


Headache

Sorafenib

VEGFR, PDGFR, Raf kinase, KIT, FLT-3

Renal cell carcinoma, hepatocellular carcinoma, radioactive iodine-refractory thyroid cancer


Ischemic stroke

Trametinib

MEK

Melanoma


Ocular toxicity (blurred vision, chorioretinopathy)

Palbociclib

CDK4, CDK6

ER+ metastatic breast cancer

Headache, dizziness, peripheral neuropathy


Lipid kinase inhibitors

Idelalisib

PI3K delta

CLL, follicular B-cell NHL, small lymphocytic lymphoma

Headache


Histone deacetylase inhibitors

Vorinostat

HDAC

Cutaneous T-cell lymphoma

Muscle spasms, headache


Belinostat

HDAC

Peripheral T-cell lymphoma

Dizziness, headache

Apraxia

Romidepsin

HDAC

Cutaneous T-cell lymphoma

Headache


Panobinostat

HDAC

Multiple myeloma

Headache, dizziness, back pain, peripheral neuropathy


mTOR inhibitors

Everolimus

mTOR

Breast cancer, pancreatic cancer, RCC, SEGA in tuberous sclerosis patients

Convulsionsa, headache


Sirolimus

mTOR

Immunosuppression in solid organ transplant, lymphangioleiomyomatosis


RPLS

Temsirolimus

mTOR

Advanced RCC


CNS hemorrhage

Smoothened inhibitors

Vismodegib

SMO

Basal Cell Carcinoma

Muscle spasms


Sonidegib

SMO

Basal Cell Carcinoma

Muscle spasms, headache


PARP inhibitors

Olaparib

PARP

Ovarian cancer

Headache, peripheral neuropathy

Hemorrhagic stroke


aIn tuberous sclerosis patients; frequency not increased in comparison with placebo

AIDP acute inflammatory demyelinating polyneuropathy, ALL acute lymphoblastic leukemia, ALK anaplastic lymphoma kinase, BCR-ABL breakpoint cluster region-Abelson murine leukemia viral oncogene homolog 1, BTK Bruton’s tyrosine kinase, CDK cyclin-dependent kinase, c-Fms colony-stimulating factor 1 receptor , CLL chronic lymphocytic leukemia, CML chronic myelogenous leukemia, CNS central nervous system, EGFR epidermal growth factor receptor, ER estrogen receptor, FLT-3 FMS-like tyrosine kinase 3, GIST gastrointestinal stromal tumor, HDAC histone deacetylase, HER human epidermal growth factor receptor, JAK Janus kinase, MDS myelodysplastic syndrome, MEK mitogen-activated protein kinase kinase, MPD Myeloproliferative disorder, mTOR mammalian target of rapamycin, NHL non-Hodgkin lymphoma, NSCLC non-small cell lung cancer, PARP poly (ADP-ribose) polymerase, PDGFR platelet-derived growth factor receptor, PI3K phosphoinositide-3-kinase, RCC renal cell carcinoma, RET REarranged during Transfection, RPLS reversible posterior leukoencephalopathy syndrome, SEGA subependymal giant cell astrocytoma, SMO smoothened, TIA transient ischemic attack,  TIE2 tunica interna endothelial cell kinase, VEGFR vascular endothelial growth factor receptor


Proteasome Inhibitors



Bortezomib


Bortezomib is a reversible inhibitor of the 26S proteasome, and is FDA approved for the treatment of multiple myeloma and patients with mantle cell lymphoma who have received at least one prior therapy [47]. The most significant neurotoxicity associated with bortezomib is treatment-emergent peripheral neuropathy. In studies of treatment-naïve multiple myeloma patients receiving bortezomib, 40–64% of patients experienced peripheral neuropathy, with 14–30% requiring dose decrease or discontinuation; it was the most common reason for drug discontinuation [48]. There is a cumulative dose effect through the first five cycles of treatment, and there has been some suggestion that the route of administration may have an impact, with one study showing significantly decreased incidence of neuropathy with subcutaneous versus intravenous administration [49]. Bortezomib typically causes a painful, length-dependent, axonal sensory neuropathy which is often at least partly reversible upon discontinuation of treatment [47]. Neurologic examination, nerve conduction studies and electromyography are often normal except in severe cases [50]. Constipation and orthostasis, noted in 10–15% of patients receiving bortezomib, are thought to be largely mediated by autonomic neuropathy. Motor nerve involvement is rare [41]. Other than neuropathy, neurologic complications are rare, although isolated cases of bortezomib-induced PRES have been reported (Fig. 16.3) [51, 52].

A328796_3_En_16_Fig3_HTML.jpg


Fig. 16.3
Bortezomib-Induced Posterior Reversible Encephalopathy Syndrome MRI of the brain in a 66-year-old woman recently treated with bortezomib for multiple myeloma, who presented with new onset of seizures. The T2/FLAIR images shown are notable for fairly symmetric cortical and subcortical hyperintense signal abnormalities with a posterior predominance (Image courtesy of Aaron Berkowitz, M.D., Ph.D., Brigham and Women’s Hospital.)


Carfilzomib


Carfilzomib is a second-generation proteasome inhibitor that is FDA approved as monotherapy for the treatment of progressive multiple myeloma in patients previously on treatment with bortezomib and an immunomodulatory agent, and in combination with lenalidomide and dexamethasone in patients with recurrent multiple myeloma previously treated with one to three other regimens. Carfilzomib has significantly less neurotoxicity than bortezomib; this is felt to be possibly explained by the off-target effects on non-proteasome serine proteases seen with bortezomib but not carfilzomib [53]. In an integrated safety analysis of four phase II clinical trials using single-agent carfilzomib, 84.8% of patients had a history of treatment-related peripheral neuropathy (from bortezomib or thalidomide), with 71.9% of patients with active peripheral neuropathy at the time of trial enrollment (all grades 1 or 2). However, there were infrequent reports of neuropathy as an adverse side effect in these trials, reported in 13.9% of patients overall, the majority of which were grades 1–2 (1.3% of patients experienced grade 3 peripheral neuropathy, all in patients with preexisting grade 1–2 neuropathy at baseline). There was no grade 4 neuropathy and as neuropathy appeared to arise early in treatment, there was no evidence of cumulative toxicity [54].


Tyrosine Kinase Inhibitors



Epidermal Growth Factor Receptor Inhibitors



Erlotinib Hydrochloride

Erlotinib is an EGFR tyrosine kinase inhibitor that is FDA approved for use as first-line treatment of EGFR mutant, metastatic non-small cell lung cancer, as well as for pretreated advanced or metastatic non-small cell lung cancer. It is also approved for use in conjunction with gemcitabine hydrochloride in the treatment of pancreatic cancer. In the original lung cancer trials, neurologic side effects were uncommon [55]. Death related to stroke was reported in a phase III trial using erlotinib in combination with gemcitabine in patients with advanced pancreatic cancer; this was ultimately attributed to a combination of cancer and treatment-related complications [56].


Gefitinib

The EGFR inhibitor gefitinib received accelerated FDA approval in 2003 for the treatment of locally advanced or metastatic non-small cell lung cancer, based on the use of tumor response rate as a surrogate endpoint for clinical efficacy in clinical trials. In 2005, after follow-up studies failed to show an overall survival benefit, its use in the United States was subsequently limited to patients who were currently or previously benefiting from its use [57]. In 2015, it received additional FDA approval for use in metastatic non-small cell lung cancer with specified epidermal growth factor receptor gene mutations [58]. It remains the more widely used EGFR tyrosine kinase inhibitor in Europe and Asia, and is under ongoing investigation in lung cancer and other malignancies [59]. Gefitinib is well tolerated from the neurologic perspective, with no significant adverse neurologic side effects [60].


Afatinib Dimaleate

Afatinib , an irreversible inhibitor of EGFR, human epidermal growth factor receptor 2 (HER2) and HER4, is FDA approved for the treatment of EGFR mutant, metastatic non-small cell lung cancer. There are no significant neurologic side effects commonly associated with afatinib therapy [61, 62]


Lapatinib Ditosylate

Lapatinib is an oral small molecule HER2 and EGFR1 tyrosine kinase inhibitor. It is FDA approved for use in combination with letrozole or capecitabine in the treatment of patients with HER2 overexpressing breast cancer. Other than headache, neurologic complications are not commonly described [63, 64].


Vandetanib

Vandetanib , an oral selective inhibitor of EGFR, RET and VEGFR2 signaling, is FDA approved for the treatment of unresectable, locally advanced, or metastatic medullary thyroid cancer. In a randomized, double-blind, phase III trial of vandetanib versus placebo in patients with locally advanced or metastatic thyroid cancer, headache was common, noted in 26% of patients, and back pain was experienced by 9% of patients receiving vandetanib (vs. 20% in the placebo arm) [65]. Serious neurologic events were similar to those seen with other anti-VEGF agents, including RPLS and an increased risk of cerebrovascular events (cerebral ischemia or transient ischemic attack were noted in 1.3% of patients in the vandetanib arm and no patients in the control arm) [66].


Anaplastic Lymphoma Kinase (ALK) Inhibitors



Crizotinib

Rearrangements of the ALK gene with the echinoderm microtubule associated protein like 4 (EML4) result in a fusion oncogene EML4-ALK which is present in 3–5% of non-small cell lung cancer. Crizotinib, a small molecule inhibitor of ALK, MET and ROS1 tyrosine kinases, is FDA approved for the treatment of ALK-rearranged non-small cell lung cancer, with clinical trials showing superiority to standard chemotherapy in patients with progressive tumors. It is generally neurologically well tolerated, although visual side effects are common and may include diplopia, photopsia, chromatopsia, blurred vision, impaired vision, and vitreous floaters. Dizziness, generally mild, occurred in about 20% of patients in clinical studies [67, 68].


Ceritinib

In April 2014, certinib, an oral, small molecule tyrosine kinase inhibitor of ALK, received accelerated FDA approval for the treatment of ALK-rearranged non-small cell lung cancer with progression on (or intolerance to) crizotinib, due to evidence of significant and durable treatment responses [69]. Neurologic adverse events (regardless of attribution to study drug) in phase I clinical trial data included headache (15%), dizziness (11%), muscle spasms (8%), dysphonia (7%), tremor (7%), and convulsion (6%) [70].


MET Inhibitors



Cabozantinib

Cabozantinib is an inhibitor of MET, VEGFR2, and RET that is FDA approved for the treatment of metastatic medullary thyroid cancer. It is associated with significant toxicity, requiring dose reduction in 79% of patients in the phase III trial. With regard to neurologic toxicity, side effects noted in 10% or more of patients included dysphonia, back pain, headache, and dizziness. RPLS was reported in one patient, and other VEGF inhibitor-associated complications were seen, including hemorrhage, venous thrombosis, and arterial thrombosis [71].


Crizotinib

Crizotinib , a small molecule inhibitor of ALK, MET and ROS1 tyrosine kinases, is FDA approved for the treatment of ALK-rearranged non-small cell lung cancer. Neurologic complications associated with this agent are discussed in greater detail earlier in this chapter.


BCR-ABL Inhibitors



Imatinib Mesylate

Imatinib is a potent inhibitor of BCR-ABL, PDGFRα, PDGFRβ, c-Fms, and c-Kit tyrosine kinases. Initially pioneered in the treatment of Philadelphia chromosome-positive chronic myelogenous leukemia (CML), it is also FDA approved for the treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia (ALL), chronic eosinophilic leukemia, dermatofibrosarcoma protuberans, gastrointestinal stromal tumor, myelodysplastic/myeloproliferative neoplasms, and systemic mastocytosis. Neurologic adverse effects include headache, which may be severe, and muscle spasms, which are generally mild and respond to treatment with quinine, calcium or magnesium [7274]. In a phase I/II study of imatinib for the treatment of recurrent malignant gliomas, central nervous system hemorrhage was observed at a rate higher than the expected spontaneous hemorrhage rate in these patients [75]. Subdural hemorrhage in association with imatinib use also has been described [74, 76].


Nilotinib

The second-generation BCR-ABL tyrosine kinase inhibitor nilotinib has a higher selectivity and binding affinity than imatinib, and is FDA approved for the treatment of newly diagnosed and previously treated patients with CML [77]. Headaches (grade 3 or 4 in some cases, at higher incidence than with imatinib) and less commonly muscle spasm have been associated with nilotinib therapy. Rare cases of CNS hemorrhage have been described [72, 73].


Dasatinib

Dasatinib is a second-generation oral BCL-ABL inhibitor that also has activity against the SRC family kinases Lyn and Src. It is used in the treatment of Philadelphia chromosome-positive ALL and Philadelphia chromosome-positive CML. [78]. Headache, generally mild, is reported in 12–27% of patients [72, 79]. There have been several reported cases of spontaneous subdural hemorrhage in patients on dasatinib treatment, including one case in the absence of thrombocytopenia; dasatinib-induced platelet dysfunction has been implicated as the putative mechanism [80, 81].


Bosutinib

Bosutinib is an oral, dual SRC/ABL1 tyrosine kinase inhibitor that is FDA approved for the treatment of patients with Philadelphia chromosome-positive CML, resistant to or intolerant of prior therapy. In long-term follow-up of the Bosutinib Efficacy and Safety in Newly Diagnosed CML trial (BELA trial), neurologic side effects included headache in 13% of bosutinib-treated patients (vs. 11% with imatinib) and muscle spasms in 4% of bosutinib-treated patients (vs. 22% of those treated with imatinib) [82].


Ponatinib

Ponatinib is an oral multi-target tyrosine kinase inhibitor designed as a pan-BCR-ABL inhibitor but also effective against VEGFR, PDGFR, FGFR, ephrin receptor, Src family kinases, c-Kit, RET, TIE2, and FLT-3 [30]. It is used in the treatment of patients with chronic myeloid leukemia (CML) or Philadelphia chromosome-positive acute lymphoblastic leukemia. Neurologic complications of this agent are described in greater detail earlier in this chapter.


Bruton’s Tyrosine Kinase (BTK) Inhibitors



Ibrutinib

Ibrutinib is an irreversible non-receptor tyrosine kinase inhibitor that targets Bruton’s tyrosine kinase, a B-cell receptor signaling component which plays a role in the pathogenesis of chronic lymphocytic leukemia [83]. It is FDA approved for use in the treatment of chronic lymphocytic leukemia, mantle cell lymphoma, and Waldenström’s macroglobulinemia. In long-term follow-up of a phase II trial of 111 mantle cell lymphoma patients treated with single-agent ibrutinib, bleeding (including bruising) occurred in 50% of patients during the total study period, the majority of which were not neurologic. Subdural hematomas were reported in four patients, all of which were associated with head trauma and/or falls, and all four patients had received anti-platelet (aspirin) or anticoagulant (warfarin) agents in close proximity to the event. Other neurologic adverse events included confusional state in 3% of patients, and back pain [84].


Janus Kinase (JAK) Inhibitors



Ruxolitinib

The potent and selective JAK 1 and 2 inhibitor ruxolitinib is FDA approved for the treatment of polycythemia vera and for the treatment of intermediate and high-risk myelofibrosis. It is well tolerated from the neurologic standpoint. Although headache, dizziness, and muscle spasms have been described with its use, the phase III trial examining ruxolitinib in comparison with standard therapy in polycythemia vera found that 49% of patients treated with ruxolitinib had an at least 50% reduction in their Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF), with decrease in hyperviscosity-related neurologic symptoms including vision problems, dizziness, concentration problems, headache, numbness or tingling in the hands or feet, and tinnitus [85, 86].


Serine/Threonine Kinase Inhibitors



BRAF Inhibitors



Vemurafenib

40–60% of cutaneous melanomas have activating mutations in BRAF at codon 600; 80–90% of these are the V600E valine to glutamic acid substitution. These mutations result in constitutive activation of the MAPK pathway and are thought to be a driver of melanoma proliferation [87, 88]. Vemurafenib is a potent BRAF inhibitor, and is FDA approved for the treatment of unresectable or metastatic melanoma with the BRAF V600E mutation. In the randomized trial which led to its approval, 675 previously untreated patients with BRAF V600E mutation positive metastatic or unresectable melanoma were randomized to treatment with vemurafenib versus dacarbazine. Headaches were common in the vemurafenib group (23.2% vs. 10.4% in the dacarbazine group). Otherwise, neurologic side effects were uncommon [89]. Rarely, there have been cases of facial palsy attributed to vemurafenib therapy [90]. There is an isolated case report of acute inflammatory demyelinating polyneuropathy (AIDP) in the setting of vemurafenib treatment, in a patient previously treated with nivolumab [91].


Dabrafenib

Dabrafenib is a reversible, competitive inhibitor of BRAF with selective inhibition of the BRAF V600E kinase. It is FDA approved alone or in conjunction with trametinib for the treatment of unresectable or metastatic melanoma with the BRAF V600E or V600 K mutation. There are no serious neurologic complications; in the phase III trial comparing dabrafenib with dacarbazine in melanoma patients, headache was noted in 5% of dabrafenib patients (vs. 0% with dacarbazine) but it was otherwise well tolerated from the neurologic standpoint [88].

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Dec 24, 2017 | Posted by in NEUROLOGY | Comments Off on Neurological Complications of Targeted Therapies

Full access? Get Clinical Tree

Get Clinical Tree app for offline access