Skip to main content
Neuro-Oncology logoLink to Neuro-Oncology
. 2017 Jun 19;20(1):24–36. doi: 10.1093/neuonc/nox115

Neurological complications of new chemotherapy agents

Alicia M Zukas 1, David Schiff 2,3,
PMCID: PMC5761528  PMID: 28992326

Abstract

This last decade has yielded more robust development of cancer treatments and first-in-class agents than ever before. Since 2006, nearly one hundred new drugs have received regulatory approval for the treatment of hematological and solid organ neoplasms. Moreover, older conventional therapies have received approval for new clinical indications and are being used in combination with these newer small-molecule targeted treatments. The nervous system is vulnerable to many of the traditional cancer therapies, manifesting both already well-described acute and chronic toxicities. However, newer agents may produce toxicities that may seem indistinguishable from the underlying cancer. Early recognition of neurotoxicities from new therapeutics is vital to avoid irreversible neurological injury. This review focuses on cancer therapies in use in the last 10 years and approved by the FDA from January 2006 through January 1, 2017.

Keywords: antineoplastic agents, central nervous system, neurotoxicity syndromes, peripheral nervous system


Chemotherapy-related neurotoxicity is a well-recognized complication encountered by cancer survivors. Chemotherapy neurotoxicity tends to relate to drug mechanism of action, dose intensity, preexisting neurological injury, or underlying systemic disease. In the last decade, the FDA Center for Drug Evaluation and Research approved nearly one hundred new molecular entities or therapeutic biologics for the treatment of cancer (Tables 1, 2, and 3). In the last 10 years, there has been an expansive development of targeted therapies, and these have introduced a yet wider range of potential toxicities. Twenty classes of receptor tyrosine kinases (RTKs) have been described, each with subclasses and potential drug targets (Table 1).1 Their small size is a convenient passage threshold for the blood–brain barrier.2 Because they are generally metabolized through cytochromes P450, discretion is necessary if used in conjunction with classical inhibitors and inducers of cytochromes P450. Several of these newer agents, including some antibody therapies (Table 2), carry risks for associated neurological disease, such as progressive multifocal leukoencephalopathy (PML), increasing the risk for vaso-occlusive events, or posterior reversible leukoencephalopathy syndrome (PRES; Fig. 1). New immunotherapies incorporate various mechanisms to unleash the immune system against the tumor, though with the risk of engaging the immune system against self and potentially provoking an autoimmune neurohypophysitis (Fig. 2), myositis, or myasthenia gravis. Neurological toxicities from treatments may seem indistinguishable from the direct effects of the cancer itself or its indirect complications. Recognition and differentiation of these complications is imperative to the proper care and treatment of the patient. In this review, we summarize the neurotoxicological profiles of the newer cancer therapies approved for clinical use in the last 10 years.

Table 1.

Small-molecule kinase inhibitors approved by the FDA in the last 10 years

Drug Trade Name Approval Year Cancer Target
Afatinib Gilotrif 2013 Metastatic NSCLC with EGFR mutation HER2, EGFR
Alectinib Alecensa 2015 ALK+ metastatic NSCLC ALK
Axitinib Inlyta 2012 RCC PDGFR, c-Kit, VEGFR, BCR-Abl
Bosutinib Bosulif 2012 Ph+ CML BCR-Abl, Src family
Cabozantinib Cometriq 2012 Metastatic medullary thyroid cancer c-MET, VEGFR
Ceritinib Zykadia 2014 ALK+ metastatic NSCLC ALK
Cobimetinib Cotellic 2015 BRAF V600E or V600K melanoma MEK
Crizotinib Xalkori 2011 ALK+ NSCLC MET, ALK, ROS1
Dabrafenib Tafinlar 2013 BRAF V600E melanoma BRAF
Dasatinib Sprycel 2006 CML with imatinib resistance BCR-Abl, Src family
Ibrutinib Imbruvica 2013 Mantle cell lymphoma, CLL BTK
Idelalisib Zydelig 2014 CLL, follicular NHL, and small lymphocytic lymphoma PI3K
Lapatinib Tykerb 2007 Breast cancer HER2, EGFR
Lenvatinib Lenvima 2015 Thyroid cancer VEGFR1, VEGFR2, VEGFR3
Nilotinib Tasigna 2007 CML BCR-Abl
Osimertinib Tagrisso 2015 EGFR T790M mutation NSCLC EGFR
Palbociclib Ibrance 2015 ER+ HER2− breast cancer CDK4, CDK6
Panitumumab Vectibix 2006 Colorectal cancer EGFR
Pazopanib Votrient 2009 Soft tissue sarcoma, RCC c-Kit, FGFR, PDGFR, VEGFR
Ponatinib Iclusig 2012 CML, Ph+ ALL BCR-Abl
Regorafenib Stivarga 2012 GIST, metastatic colorectal cancer VEGFR2
Sunitinib Sutent 2006 RCC, GIST, pancreatic NET VEGFR, PDGFR, c-Kit, RET
Trametinib Mekinist 2013 BRAF V600E or V600K melanoma MEK
Vandetanib Vandetanib 2011 Thyroid cancer VEGFR, RET, EGFR
Vemurafenib Zelboraf 2011 BRAF+ melanoma BRAF

Abbreviations: RCC, renal cell carcinoma; NHL, non-Hodgkin lymphoma; GIST, gastrointestinal stromal tumor; NET, neuroendocrine tumor.

Table 3.

Additional cancer therapies recently approved, their trade names, approval year, cancer, and mechanism

Drug Trade Name Approval Year Cancer Mechanism
Abiraterone Zytiga 2011 Prostate cancer Anti-androgen
Belinostat Beleodaq 2014 Relapsed T-cell lymphoma HDAC inhibitor
Cabazitaxel Jevtana 2010 Prostate cancer Antimicrotubule, taxane derivative
Carfilzomib Kyprolis 2012 Multiple myeloma Proteasome inhibitor
Degarelix Degarelix 2008 Prostate cancer Anti–gonadotropin-releasing hormone
Enzalutamide Xtandi 2012 Prostate cancer Anti-androgen
Eribulin mesylate Halaven 2010 Metastatic breast cancer Antimicrotubule
Everolimus Afinitor 2009 Renal angiomyolipoma, SEGA, breast cancer, NET, RCC mTOR kinase inhibitor
Ixabepilone Ixempra 2007 Breast cancer Antimicrotubule, taxane derivative
Ixazomib Ninlaro 2015 Multiple myeloma Proteasome inhibitor
Ms-275 Entinostat 2013 Breast cancer HDAC inhibitor
Olaparib Lynparza 2014 BRCA mutated advanced ovarian cancer PARP inhibitor
Omacetaxine mepesuccinate Synribo 2012 CML Cephalotaxine, protein synthesis inhibitor
Pomalidomide Pomalyst 2013 Multiple myeloma Thalidomide analog
Pralatrexate injection Folotyn 2009 Peripheral T-cell lymphoma Antimetabolite
Romidepsin Istodax 2009 CTCL HDAC inhibitor
Rucaparib Rubraca 2016 BRCA mutated advanced ovarian cancer PARP inhibitor
Sipuleucel-T Provenge 2010 Prostate cancer CD54+ cells, autologous
Sonidegib Odomzo 2015 Advance basal cell carcinoma Hedgehog pathway inhibitor
Talimogene laherparepvec Imlygic 2015 Recurrent melanoma Oncolytic virus
Temsirolimus Torisel 2007 RCC mTOR kinase inhibitor
Trabectedin Yondelis 2015 Liposarcoma or leiomyosarcoma FUS-CHOP inhibitor
Trifluridine and tipiracil Lonsurf 2015 Metastatic colorectal cancer Antimetabolite
Venetoclax Venclexta 2016 CLL with 17p deletion Bcl-2 inhibitor
Vincristine sulfate liposomes Marqibo 2012 ALL Antimicrotubule, vinca alkaloid
Vismodegib Erivedge 2012 Basal cell carcinoma Hedgehog pathway inhibitor
Vorinostat Zolinza 2006 CTCL HDAC inhibitor

Additional abbreviations: CTCL, cutaneous T-cell lymphoma; SEGA, subependymal giant cell astrocytoma.

Table 2.

Antibody therapies recently approved by the FDA, their trade names, approval year, cancers for which they are approved, and their targets

Drug Trade Name Approval Year Cancer Antibody Target
Blinatumomab Blincyto 2014 Ph− B cell ALL CD19/CD3
Obinutuzumab Gazyva 2013 CLL CD20
Ofatumumab Arzerra 2009 CLL CD20
Brentuximab vedotin Adcetris 2011 Hodgkin lymphoma, large cell lymphoma CD30
Daratumumab Darzalex 2015 Multiple myeloma CD38
Ipilimumab Yervoy 2011 Metastatic melanoma CTLA-4
Necitumumab Portrazza 2015 Metastatic squamous NSCLC EGFR
Dinutuximab Unituxin 2015 Pediatric neuroblastoma GD2
Pertuzumab Perjeta 2012 HER2+ metastatic breast cancer HER2
Ado-trastuzumab emtansine Kadcyla 2013 HER2+ metastatic breast cancer HER2, antimicrotubule agent
Nivolumab Opdivo 2015 Metastatic squamous NSCLC, metastatic melanoma PD-1
Pembrolizumab Keytruda 2014 Unresectable, recurrent, and metastatic melanoma PD-1
Atezolizumab Tecentriq 2016 NSCLC, urothelial carcinoma PD-1 ligand
Olaratumab Lartruvo 2016 Soft tissue sarcoma PDGFRα
Elotuzumab Empliciti 2015 Multiple myeloma SLAMF7
Ziv-aflibercept Zaltrap 2012 Metastatic colorectal cancer VEGF
Ramucirumab Cyramza 2014 Gastric cancer VEGFR2

Fig. 1.

Fig. 1

Neurotoxicities implicated with their cancer therapy targets within the neuraxis.

Fig. 2.

Fig. 2

Neurohypophysitis with infundibulo-panhypophysitis induced by ipilimumab; arrow points to enhancing pituitary gland and stalk on a T1 post-gadolinium midline sagittal MRI.

Small-Molecule Kinase Inhibitors

Breakpoint Cluster Region–Abelson Murine Leukemia Kinase Inhibitors

Imatinib is the oldest of the tyrosine kinase inhibitors (TKIs), first approved in 2001, and now has 17 indications for use. Second-generation bosutinib has since emerged and been suggested to increase stroke risk, though the relative stroke risk against imatinib is uncertain. Three of the new-generation breakpoint cluster region–Abelson murine leukemia (BCR-Abl) TKIs (nilotinib, ponatinib, and dasatinib) demonstrate a moderately elevated risk of vascular occlusive events compared with imatinib.3 Nilotinib is a potent selective BCR-Abl kinase inhibitor that received FDA approval in 2007, and subsequent meta-data have indicated an increased risk of stroke, possibly via accelerating intra- and extracranial atherosclerosis or by potentiating a prothrombotic state.4,5 Ponatinib has broad-spectrum efficacy against both mutated and nonmutated BCR-Abl1 and is the most effective known drug against specifically the BCR-Abl T315I kinase domain mutation. It was issued an FDA black box warning in 2012 for arterial and peripheral vascular complications noted in 24%–48% of patients.6 The mechanism for the induced vascular damage is unclear, though it may be attributed to being both pro-atherogenic as well as disabling to plaque-stabilizing endothelial cells. Lipid and blood glucose levels should be monitored while on therapy and may lead to long-term increased risk for stroke. Dasatinib is an Src/Abl TKI approved for patients with chronic myeloid leukemia (CML) refractory to imatinib. The most common side effect of dasatinib is headache, affecting at least one-third of patients, though it has also been associated with a risk for arterial occlusive events.7 Dasatinib has also been rarely implicated with a secondary effect of optic neuropathy causing visual field loss.8

Epidermal Growth Factor Receptor Inhibitors

The human epidermal growth factor receptor (HER), of the RTK superfamily, comprises epidermal growth factor receptor (EGFR; ErbB-1), HER2 (ErbB-2), HER3 (ErbB-3), and HER4 (ErbB-4). Afatinib, erlotinib, and gefitinib are the orally available EGFR inhibitors. Cetuximab and panitumumab are intravenously administered monoclonal antibodies against EGFR.9 Erlotinib and gefitinib are reversible EGFR TKIs used in metastatic non–small cell lung carcinoma (NSCLC) harboring EGFR mutations.10 Osimertinib is a third-generation EGFR TKI used in NSCLC. Fatigue is the most common neurological complaint of the EGFR inhibitors, affecting up to 90% of patients, and more likely when used in combination therapy.11 Targeted therapies have become standard of care against metastatic breast cancer with HER2, a tyrosine kinase receptor overexpressed in 20%–30% of breast cancers. Patients with a good performance status are typically offered combination therapy of pertuzumab-trastuzumab and a taxane. Pertuzumab is a monoclonal antibody against the extracellular domain of HER2 dimerizing with other HER family members, while trastuzumab targets only HER2 homodimerization.12,13 Trastuzumab emtansine (T-DM1) is a targeted antibody-drug conjugate of DM1, a maytansine derivative that inhibits microtubule polymerization with greater potency than the vinca alkaloids. There may be significant neurotoxicity, with 2.4% having grade ≥3 peripheral neuropathy compared with 0.2% in the lapatinib plus capecitabine group in the EMILIA trial.14,15 While the brain is often the first site of metastasis in these patients, the blood–brain barrier is not penetrated well by pertuzumab and trastuzumab because of their large molecular weights. Lapatinib, approved in 2007, is a small-molecular inhibitor of HER2. Despite known CNS penetration, serious neurotoxicity remains uncommon, with 20%–41% reporting grade 1 headache per the Common Terminology Criteria for adverse events, as applied by the Common Terminology Criteria for Adverse Events.16

EGFR has also been an attractive target for inhibition by antibody therapy. Panitumumab is a human monoclonal antibody against EGFR, approved in 2006 for use against metastatic colorectal cancer, most effectively with Kras wild-type.17 With a side effect profile similar to other EGFR inhibitors, fatigue and malaise are the most common neurological complaints of these monoclonal antibodies. There have been at least 10 case reports of cetuximab-associated self-limiting aseptic meningitis occurring several hours after infusion.18 Necitumumab is an EGFR antibody used in combination therapy for metastatic squamous cell carcinoma. In the INSPIRE trial comparing necitumumab plus pemetrexed and cisplatin versus pemetrexed and cisplatin, there were greater arterial and venous thromboembolic events in the patients exposed to necitumumab without survival benefit.19

Multiple Receptor Tyrosine Kinases

Many of the vascular endothelial growth factor (VEGF) targeting agents have a similar side effect profile of fatigue, hypertension, thrombotic events, and risk for PRES. Sunitinib, FDA approved in 2006, is a multiple receptor TKI including targets VEGF receptor (VEGFR), RET (“rearranged during transfection”), and platelet-derived growth factor receptor (PDGFR). It is used in the treatment of gastrointestinal stromal tumor after progression on imatinib, renal cell carcinoma, and pancreatic neuroendocrine tumors. As targeted PDGFR antibody therapies have fairly low direct neurotoxicity, the effects may be more attributable to sunitinib’s VEGFR activity. There have been case reports of Guillain-Barré syndrome associated with sunitinib.20,21 Neurological autoimmunity is thought to be a result of VEGFR inhibition leading to increased levels of both cerebrospinal fluid (CSF) and circulating VEGF, and this may destabilize the microvasculature at the choroid plexus. There, the excess VEGF enhances angiogenesis and increases local vascular permeability, and these increased VEGF levels are comparable to those seen in Guillain-Barré and chronic inflammatory demyelinating polyneuropathy.20 The precise role of sunitinib in the primary immune response, however, has been debated and is not well understood. PRES has indeed been described in patients receiving sunitinib.22 The first TKI approved for metastatic medullary thyroid cancer, vandetanib, inhibits EGFR, VEGFR, and RET tyrosine kinase. Vandetanib has also been implicated in PRES in the context of clinical trials for primary brain tumors.23,24 Axitinib, approved in 2012, is a potent second-generation VEGF inhibitor used in renal cell cancer, also carrying risk for PRES.25–27 Lenvatinib is a multiple receptor kinase inhibitor against receptors, including VEGFR, fibroblast growth factor receptor (FGFR), PDGFR, RET, and c-Kit, and is used in progressive thyroid cancers or in combination with everolimus for renal cell carcinoma. It may also carry a similar side effect profile and appears to carry lower direct neurotoxicity than sunitinib.28 Pazopanib is a multitargeted receptor TKI against c-Kit, FGFR, PDGFR, and VEGFR used in the treatment of renal cell carcinoma and soft tissue sarcoma. There is a reported rare toxicity of hallucinations associated with its use as well as PRES.29,30 Regorafenib is an oral multikinase agent against VEGFR1/3, PDGFR, FGFR, tunica interna endothelial cell kinase 2, c-Kit, RET, Raf1, and B-Raf. It was first FDA approved in 2012 for colorectal cancer, also with low direct neurotoxicity, though with potential for VEGF target side effects.

Chronic lymphocytic leukemia (CLL) is typically treated with a combination of fludarabine, cyclophosphamide, and rituximab. Older and frail individuals with CLL or recurrent disease may not be good candidates for myelosuppressive therapy; therefore, newer agents have become attractive options for targeting the B-cell receptor pathway. Ibrutinib is a first-in-class oral irreversible inhibitor of Bruton tyrosine kinase (BTK), thereby downregulating nuclear factor-kappaB as well as several other kinases, including EGFR and Tec, as well as extracellular signal-regulated kinase (ERK).31 The side effects of BTK are typically mild; however, given its multiple targets, several side effects have been evident. In phase I/II studies for mantle cell lymphoma, 2% of patients had intracranial hemorrhage despite platelets being normal or slightly reduced.32 BTK is present on platelets, and bleeding is thought to be related to ibrutinib’s inhibition of collagen-dependent platelet aggregation.33 Ibrutinib can cross the blood–brain barrier, with up to 7% of systemic dose found in CSF.34 The most common CNS side effects are fatigue, dizziness, and headaches.35 There has also been a report of PML after ibrutinib therapy for CLL.36 Idelalisib is a first-in-class inhibitor of phosphatidylinositol 3-kinase (PI3K) delta that blocks BCR signaling as well as interrupting the homing of CLL cells from the lymph node. The PI3K inhibitors as a class are tolerated well neurologically and do not cross a healthy blood–brain barrier when studied in animals.37 They do convey a high toxicity profile systemically with a black box warning for diarrhea/colitis, hepatotoxicity, pneumonitis, and intestinal perforation.

Anaplastic lymphoma kinase (ALK) inhibitors

Crizotinib has offered improved progression-free survival in patients with ALK-rearranged NSCLC, though with 40% experiencing metastatic progression to the CNS. CNS drug side effects are largely limited to fatigue and headache. There have been rare case reports of optic neuropathy associated with crizotinib.38 Myalgias were present in almost a fifth of patients treated with alectinib, a more potent ALK inhibitor in ALK-rearranged NSCLC, with good CNS penetration and a CNS objective response rate of 57% in patients with baseline measurable CNS metastases.39 Ceritinib is an alternative for patients with progression while on crizotinib.40,41 Fatigue and neuropathy have been reported with its use, with neuropathy affecting 17% of patients.42,43 Cabozantinib is a small-molecule inhibitor targeted against both c-MET and VEGFR2 for the treatment of medullary thyroid cancer and renal cell carcinoma. Cabozantinib may overcome crizotinib resistance in ROS1 fusion acquired mutations in NSCLC; blood pressure monitoring is recommended because of risk for hypertensive crisis and PRES.44,45

BRAF Kinase Inhibitors

For the subset of patients with melanoma containing the BRAF V600 mutations, vemurafenib and dabrafenib are orally administered inhibitors of activated BRAF kinase. BRAF inhibitor therapies are radiation sensitizers, and consensus guidelines recommend avoiding its use several days prior to and after radiation in order to limit skin toxicity within the field in conventional fractionated radiotherapy.46,47 Stereotactic radiotherapy with concurrent BRAF inhibitors for melanoma may pose a risk of cerebral edema and hemorrhage; overall, severe cerebral edema was seen in 15% of patients.48 There have been several reported cases of bilateral facial nerve palsy associated with vemurafenib use.49,50 The presumed mechanism is unclear but presumed to be immune mediated, as steroids appear to resolve symptoms.

MEK Inhibitors

Trametinib and cobimetinib are oral mitogen-activated protein kinase kinase (MEK) inhibitors used in melanoma with BRAF V600 mutations. They are used in combination therapy alongside BRAF inhibition.51 Patients receiving combination therapy may experience pyrexia, alleviated by low-dose corticosteroids. Elevated creatine phosphokinase is not uncommon. Rhabdomyolysis and retinal detachment with a serous retinopathy are class effects of MEK inhibitors, affecting about 1% of patients.52

Cyclin-Dependent Kinase Inhibitor

Palbociclib is a small-molecule inhibitor of cyclin-dependent kinases 4 and 6 used in advanced hormone receptor– positive patients. Mild neurotoxicities have been reported in its use, such as myalgia, fatigue, and dysgeusia.53

Other Novel Inhibitors

Hedgehog Pathway Inhibitors

Sonidegib and vismodegib are Smoothened antagonists targeting the hedgehog pathway for the treatment of basal cell carcinomas. Both sonidegib and vismodegib have class-specific neurotoxicities of dysgeusia, myalgia, and muscle spasms.54 The hedgehog pathway is known to have activity in the taste papillae, and dysgeusia rates are as high as 57% in studies using vismodegib.55 Zinc gluconate supplementation may help in dysgeusia management.

Histone Deacetylase Inhibitors

While histone deacetylase (HDAC) compounds have several different classifications of chemical structures, they possess a similar toxicity profile. Vorinostat, belinostat, and panobinostat are all hydroxamic acids, while romidepsin is a depsipeptide, and entinostat is a benzamide. All have activity against class 1 HDAC enzymes but different activities against other HDAC isoenzymes. Fatigue is a common constitutional symptom seen in all HDAC inhibitors, with dose-limiting toxicity in a quarter of patients in phase II trials.56

Mechanistic TOR Kinase Inhibitors

The first generation of mechanistic target of rapamycin (mTOR) inhibitors are the rapalogs, including rapamycin, everolimus, and temsirolimus, with forthcoming second- and third-generation agents. The rapalogs share a common class effect, and while these may be very severe to other organ systems, the rate of direct neurological side effects is rare. Rapamycin treatment has been attributed to the development of PRES in organ transplant patients.57 Rapalogs induce hyperglycemia and an mTOR-induced diabetes, and there is an mTOR-induced dyslipidemia which may elevate risk for stroke.58 Phosphate levels may be reduced, which may manifest as fatigue and muscle weakness.

PARP Inhibitors

Olaparib and rucaparib are oral poly(ADP-ribose) polymerase (PARP) inhibitors for the treatment of ovarian cancer in patients with breast cancer (BRCA) mutation and prior chemotherapy. Neurotoxicity is minimal, with the most common toxicity being fatigue.59 Nearly half of patients on rucaparib had mild dysgeusia.60

Protein Synthesis Inhibitors

Omacetaxine mepesuccinate, formally homoharringtonine, is a subcutaneously delivered protein synthesis inhibitor intended for patients with CML with intolerance to 2 or more TKIs. It received accelerated FDA approval in 2012, and safety data revealed significant hematological toxicity, with high-grade thrombocytopenia occurring in 76% of patients and reported deaths from intracerebral hemorrhage.61

Proteasome Inhibitors

Second-generation proteasome inhibitors are now approved for treatment of multiple myeloma. Carfilzomib and ixazomib have demonstrated less painful sensory neuropathy than bortezomib, which tends to carry a high rate of neuropathy.62,63 The CIPN is typically reversible with drug discontinuation or dose reduction, though this may take months to years. Ixazomib is a once weekly fixed dose oral therapy and is considered to carry the lowest incidence of CIPN.64 As it is used now, the sensory neuropathy reported is typically grade 1 or 2 and this is likely related to prior exposure to bortezomib.65

B-cell Lymphoma 2 Inhibitor

Venetoclax is a first-in-class B-cell lymphoma (Bcl-2) inhibitor newly approved for use in hematological malignancies. Despite significant hematological toxicity, neurotoxicity is limited to low-grade fatigue (40%) and headache (24%).66

Antibody Therapies

Anti-CD20 Antibodies

Postmarketing data of rituximab found that 22 patients developed PML in 2006; in 2007, the FDA issued a black box warning for all anti-CD20 antibodies. Ofatumumab is a human type I CD20 monoclonal antibody FDA approved in 2014 for second-line treatment of CLL. Obinutuzumab is a type II monoclonal antibody against CD20 approved in 2013 and is used against CLL and follicular lymphoma. Obinutuzumab provokes more direct cell-mediated cytotoxicity than the type I antibodies, which tend to be stronger activators of complement-mediated cytotoxicity. Obinutuzumab has been implicated in an increased risk for PML.67 Patients receiving monoclonal antibody treatment may present with different clinical features from those presenting with classic PML, such as sensory changes and seizures.68 Definitive diagnosis, according to 2013 guidelines, is made by confirming the presence of John Cunningham (JC) virus in CSF by PCR, or brain biopsy if JC virus is undetectable in CSF and alternative explanations are unrevealing.69

Vascular Endothelial Growth Factor Antibodies

Bevacizumab is a monoclonal immunoglobulin (Ig)G1 antibody against human VEGF with multiple clinical applications and a shared side effect profile with other anti-angiogenic agents against VEGF and its receptor. As a class, inhibitors against VEGF include hypertension, proteinuria, and delayed wound healing. The anti-VEGF-mediated hypertension may be a product of nitric oxide pathway inhibition and oxidative stress.70 Thromboembolic events have been reported, and in 2006, bevacizumab label was updated to indicate increased risk of PRES.71,72 Ziv-aflibercept is a fusion protein of IgG1 fused to binding portions of VEGFR used in metastatic colorectal cancer. Both arterial and venous thromboembolism have been seen with its use, likely due to its effects on VEGF. In a phase III study, thromboembolic events were seen in 9.7% of the treatment arm compared with 6.8% in the control arm.73 Ramucirumab is a monoclonal antibody against the extracellular domain of VEGFR2 and is used as salvage therapy in gastric and colorectal cancer. It carries similar toxicities as the aforementioned anti-angiogenic agents.74

Platelet-Derived Growth Factor Receptor Antibodies

Olaratumab, targeted against PDGFR alpha, was approved in late 2016 for use in soft tissue sarcoma in combination with doxorubicin. Fatigue affects 69% of patients in both the olaratumab and doxorubicin cohorts.75

Anti-Glycolipid Antibodies

Dinutuximab, also referred to as ch14.18, is an anti-glycolipid disialoganglioside (GD2) monoclonal antibody used in combination with cytokines, isotretinoin, and granulocyte-macrophage colony-stimulating factor for the maintenance treatment of high-risk pediatric neuroblastoma. Gangliosides are glycosylated lipid molecules, and low levels of GD2 are found on the surface of normal neurons and peripheral nerve fibers. There are fairly high GD2 levels on the cell surface of neuroblastoma, and dinutuximab triggers an antibody-dependent cell-mediated cytotoxity when it binds to GD2.76 Severe neuropathic pain, more so abdominal than in the extremities, has been seen in the first cycle, then typically dissipates in subsequent cycles, and is typically opioid resistant. Because of the pain experienced, the package insert warns against neuropathy, and the FDA has issued a boxed warning for neuropathic pain with recommendations to palliate pain, including pretreatment with acetaminophen and opiates.

Other Monoclonal Antibodies

Daratumumab is a first-in-class anti-CD38 monoclonal antibody used in multiple myeloma with no significant neurotoxicity aside from mild fatigue.77 Elotuzumab, also used against multiple myeloma, targets signaling lymphocytic activation molecule F7 (SLAMF7), which is minimally present on normal healthy tissues. The most common neurotoxicity is fatigue, affecting half of patients receiving it, and muscle spasms affecting 30%.78 Blinatumomab is a monoclonal antibody that engages CD3-CD19 in hematological malignancies. Due to its robust immune activation, it carries a black box warning for cytokine release syndrome and neurotoxicity.79 These toxicities are dose dependent, and 13%–22% of patients on blinatumomab have grade 3 or greater neurological adverse events.80,81 Blinatumomab releases pro-inflammatory cytokines which peak 2–48 hours after the first dose.82 Neurological symptoms are likely due to pro-inflammatory cytokines at the neuroendothelium.83 Patients may present with seizures, encephalopathy, coma, aphasia, or imbalance occurring about a week after therapy. Neurotoxicity prophylaxis involves premedication with dexamethasone. In a phase II BiTE trial, a fifth of patients had grade 3 neurotoxicity, most commonly encephalopathy, somnolence, and disorientation. Symptoms were treated with dexamethasone 8 mg i.v. every 8 hours for 3 days followed by a taper with complete resolution in all but one patient in each cohort.84

Immunotherapies

The immune system plays a pivotal role in recognizing and preventing the growth of cells with tumorigenic potential, and tumors may hold immune resistance through altered immune checkpoint pathways. Checkpoint molecules include the newly targeted programmed cell death-1 (PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4). The treatment for metastatic melanoma, in particular, has made substantial progress in recent years with the use of checkpoint inhibitors.85 Common side effects include mild fatigue and headache, with grade 3 cases affecting less than 3% of patients. Autoimmune toxicities from checkpoint inhibitors are typically distinct from cytokine therapy toxicities, with a majority of the events involving the skin, gastrointestinal tract, and the endocrine system, including hypophysitis and thyroid disorders.86 Typically, an autoimmune adverse event occurs within the first 3 months of therapy. The use of steroids to mitigate autoimmune toxicity has not been shown to affect the overall efficacy of the therapy.87 Both the CNS and peripheral nervous system have been vulnerable to autoimmunity. CNS demyelination has been reported in conjunction with checkpoint inhibitors for several cases. One case report reveals a patient on ipilimumab, a CTLA-4 inhibitor, with both acute optic neuritis and focal CNS neurological deficits complementary to the demyelination seen on MRI and later confirmed by biopsy.88 Another patient had a severe multiple sclerosis relapse after being treated with ipilimumab for melanoma.89 A metastatic melanoma patient being treated with nivolumab, an IgG4 anti–PD-1 monoclonal antibody, had a demyelinated tumefactive lesion with extensive CNS demyelination on autopsy in the absence of CNS metastases.90 Pembrolizumab also targets PD-1 and is used in treating metastatic melanoma. There are rare cases of peripheral inflammatory neuropathy, Guillain-Barré syndrome, and myasthenia gravis with the application of these agents.91 Both sensory and motor immune-mediated polyneuropathies occur in less than 1% of patients.92 Typically the neuropathic symptoms are mild and steroid responsive and do not require discontinuation of therapy. There are rare cases of more severe and refractory steroid-responsive polyradiculitis and meningoradiculitis while on checkpoint inhibitors.93 Meningoradiculitis was diagnosed in a patient on anti–PD-1 therapy with proximal arm pain with weakness which was responsive to oral steroids but recurrent when steroids were discontinued.90 Another patient from the same series presented with leg weakness and paresthesia, with an MRI revealing contrast enhancement of the caudal nerve fibers. The patient’s immune-mediated polyradiculitis was diagnosed and treated with steroids and his checkpoint inhibitor was discontinued, but he subsequently developed progressive disease. A demyelinating polyradiculopathy has also been implicated with anti–PD-1 therapy.94 There have been a dozen case reports of newly diagnosed seropositive and seronegative myasthenia gravis while on checkpoint inhibitors.95 These cases widely vary in their patient outcomes to treatment regimens of steroids, plasmapheresis, intravenous immunoglobulin, or a combination. Inflammatory myopathy, dermatomyositis, and myositis have also been described. Dermatomyositis was described in a patient receiving ipilimumab, presenting with a cutaneous eruption followed by acute proximal muscle weakness and a significantly elevated creatine kinase and aldolase.96 Ipilimumab was discontinued and the dermatomyositis was responsive to high-dose steroids. Early recognition of neurotoxicities may mitigate their severity, with corticosteroids being the primary therapy for immune-oncology drug-related reactions. Low-grade toxicities may be treated with oral steroid dosing strategies, while more severe toxicities may be addressed with intravenous steroids, with more severe cases even considering i.v. Ig.91

Hypophysitis is a known toxicity of both anti–CTLA-4 and PD-1 inhibition and may occur in up to 5% of patients.97 It may present as a variety of neurological symptoms, including fatigue and headache. Hypophysitis is diagnosed by measuring low pituitary hormone levels, and a brain MRI may show enhancement or enlargement of pituitary or its infundibulum (Fig. 2). Treatment typically consists of high-dose steroids and hormone supplementation. Atezolizumab is a PD-1 ligand inhibitor approved in 2016 for urothelial carcinoma and NSCLC. No serious neurological immune-related adverse events have yet been described in its phase II trials (BIRCH, POPLAR, FIR).98

Classical Therapies

Antimicrotubule Agents

Chemotherapy-induced peripheral neuropathy (CIPN) is a familiar side effect of many conventional vinca alkaloids, cisplatin, taxanes, and thalidomide agents. In 2012, vincristine emerged in a sphingomyelin/cholesterol encapsulated form (VSLI) for the treatment of relapsed Philadelphia chromosome negative acute lymphoblastic leukemia (ALL).99 This formula was designed for improved drug exposure to the tumor while reducing systemic toxicity. Peripheral neuropathy remains the most common dose-related nonhematological toxicity, with 10% requiring discontinuation of the therapy.100 In one study of ALL patients receiving VSLI at 2.25 mg/m2, patients could tolerate higher dosing with no reported grade 3 peripheral motor neuropathy and lower rates of neuropathy and paresthesia than what is reported for patients receiving standard vincristine at 1.4 mg/m2.101 CIPN can also be a dose-limiting toxicity with several newer microtubule-binding agents—eribulin mesylate, cabazitaxel, and ixabepilone. Within this class, paclitaxel and ixabepilone have highest risk for high-grade neuropathy, while eribulin conveys a lower risk despite higher dorsal root ganglion penetration.102 Optic neuropathy has also been reported in both older taxanes as well as cabazitaxel, approved since 2010 for hormone-refractory prostate cancer.103 Overall, there is a lower frequency and grade of both neuropathy and dysgeusia in prostate cancer patients receiving cabazitaxel compared with docetaxel.104

Brentuximab vedotin is an anti-CD30 antibody-drug conjugate approved for the treatment of relapsed Hodgkin lymphoma.105 Its vinca-like action on axonal transport is likely the mechanism of the peripheral neuropathy associated with it, though the neuropathy appears to be dose dependent and usually resolves after discontinuation of the drug.106 Additionally, its action against CD30 has implicated it in the reactivation of the JC virus leading to PML.

Antimetabolites

Since 2009 a potent intravenous folate analog pralatrexate has come to market with a milder toxicity profile than methotrexate and is currently in use for refractory T-cell lymphomas. Pretreatment with folic acid and vitamin B12 injections mitigate toxicity, and its potential efficacy is being explored in clinical trials for other lymphoid malignancies.107 Trifluridine/tipiracil is an oral antimetabolite consisting of a thymidine-based nucleoside analog with a thymidine phosphorylase inhibitor. It was FDA approved in 2015 for the treatment of metastatic colorectal cancer and carries low direct neurotoxicity. In the RECOURSE trial, while hematological adverse events were significant, less than 40% of patients reported fatigue and <5% had greater than grade 2 nonhematological toxicities.108

DNA-Damaging Agents

Trabectedin is approved for use in liposarcoma and leiomyosarcoma previously treated with anthracyclines. It is a tetrahydroisoquinoline with multiple mechanisms of action, including direct binding to DNA leading to strand breaks and inhibiting tumor angiogenesis in the microenvironment.109 While there is significant hematological and hepatic toxicity, no direct neurotoxicity has been reported in clinical trials.110

Immunomodulatory Imide Drugs

There has been further development of thalidomide analogs for myeloma and lymphoma, namely lenalidomide for mantle cell lymphoma and pomalidomide for multiple myeloma and myelofibrosis. Historically developed as a sedative, the most common neurological side effect of thalidomide and its analogs is somnolence. However, the analogs are reported to have less central and peripheral neurotoxicity than thalidomide.111 While a axonal sensory neuropathy is clinically present in up to 50% of patients, neuropathy can also be mild or subclinical, only detectable by electromyography.112,113

Anti-Androgen Therapy

Abiraterone is an anti-androgen therapy used in the treatment of prostate cancer. Fatigue is the most common severe neurotoxicity, occurring in 9% in its landmark trial.114 Enzalutamide is a second-generation androgen receptor antagonist. The AFFIRM trial reported a high risk for fatigue, and rare incidences of seizure were also reported.115 The subsequent PREVAIL trial excluded patients with history of seizure disorder. Degarelix is a gonadotropin-releasing hormone antagonist with mild neurological symptoms of fatigue not requiring discontinuation of treatment.116

Future Directions

Manipulating the immune system has been an appealing mechanism against malignancy, with recent improved survival and disease control in metastatic melanoma using ipilimumab. There are over 500 open immunotherapy clinical trials in the USA at the time of this review. Sipuleucel-T is an autologous cellular immunotherapy for the treatment of metastatic prostate cancer. It was FDA approved in 2010, and in clinical trial may present a slightly elevated risk for cerebrovascular events.117 Talimogene laherparepvec is the first oncolytic viral therapy approved in the USA, and it is injected directly into melanoma lesions with the intention to create a local antitumor response.118 In the future we may look forward to chimeric antigen receptor (CAR)–modified T-cell therapies, which are retargeted T cells used in adoptive immunotherapy.119 Their chimeric receptors are composed of a specific antigen binding domain and an intracellular T-cell activation domain. This allows an immunocompetent T cell to be redirected against a new antigen such as a tumor protein, DNA, or other biological material.120 Ideally, these will have a high specificity for the target and low long-term immunity against off-target host tissue. In clinical trials of patients receiving CAR T-cell infusions, there have been several case reports of what appears to be a cytokine release syndrome with neurological toxicity, similar to that of blinatumomab.121 Patients are described as having delirium, EEG confirmed seizures, and altered mentation requiring airway support with mechanical ventilation. CAR T cells have been found in the CSF of some of these patients by quantitative PCR.122 Reversible neurological toxicity uses CD19-CAR T cells found in CSF, with one reversible splenial lesion syndrome.123 Tocilizumab, an antibody that blocks interleukin-6 receptor, has been used to mitigate cytokine storm.124

Funding

No funding sources.

Conflict of interest statement. No conflicts of interest.

Acknowledgments

The authors would like to acknowledge the assistance of Dr Roxanne N. Sholevar for preparation of Fig. 1.

References

  • 1. Wu P, Nielsen TE, Clausen MH. FDA-approved small-molecule kinase inhibitors. Trends Pharmacol Sci. 2015;36(7):422–439. [DOI] [PubMed] [Google Scholar]
  • 2. Pardridge WM. The blood-brain barrier: bottleneck in brain drug development. NeuroRx. 2005;2(1):3–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Haguet H, Douxfils J, Mullier F, Chatelain C, Graux C, Dogné JM. Risk of arterial and venous occlusive events in chronic myeloid leukemia patients treated with new generation BCR-ABL tyrosine kinase inhibitors: a systematic review and meta-analysis. Expert Opin Drug Saf. 2017;16(1):5–12. [DOI] [PubMed] [Google Scholar]
  • 4. Alhawiti N, Burbury KL, Kwa FA et al. The tyrosine kinase inhibitor, nilotinib potentiates a prothrombotic state. Thromb Res. 2016;145:54–64. [DOI] [PubMed] [Google Scholar]
  • 5. Coon EA, Zalewski NL, Hoffman EM, Tefferi A, Flemming KD. Nilotinib treatment-associated cerebrovascular disease and stroke. Am J Hematol. 2013;88(6):534–535. [DOI] [PubMed] [Google Scholar]
  • 6. Gainor JF, Chabner BA. Ponatinib: accelerated disapproval. Oncologist. 2015;20(8):847–848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Abbas R, Hsyu PH. Clinical pharmacokinetics and pharmacodynamics of bosutinib. Clin Pharmacokinet. 2016;55(10):1191–1204. [DOI] [PubMed] [Google Scholar]
  • 8. Monge KS, Gálvez-Ruiz A, Alvárez-Carrón A, Quijada C, Matheu A. Optic neuropathy secondary to dasatinib in the treatment of a chronic myeloid leukemia case. Saudi J Ophthalmol. 2015;29(3):227–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Chakraborty S, Filippi CG, Wong T et al. Superselective intraarterial cerebral infusion of cetuximab after osmotic blood/brain barrier disruption for recurrent malignant glioma: phase I study. J Neurooncol. 2016;128(3):405–415. [DOI] [PubMed] [Google Scholar]
  • 10. Köhler J, Schuler M. Afatinib, erlotinib and gefitinib in the first-line therapy of EGFR mutation-positive lung adenocarcinoma: a review. Onkologie. 2013;36(9):510–518. [DOI] [PubMed] [Google Scholar]
  • 11. Moore MJ, Goldstein D, Hamm J et al. ; National Cancer Institute of Canada Clinical Trials Group. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25(15):1960–1966. [DOI] [PubMed] [Google Scholar]
  • 12. Adams CW, Allison DE, Flagella K et al. Humanization of a recombinant monoclonal antibody to produce a therapeutic HER dimerization inhibitor, pertuzumab. Cancer Immunol Immunother. 2006;55(6):717–727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Pickl M, Ries CH. Comparison of 3D and 2D tumor models reveals enhanced HER2 activation in 3D associated with an increased response to trastuzumab. Oncogene. 2009;28(3):461–468. [DOI] [PubMed] [Google Scholar]
  • 14. Corrigan PA, Cicci TA, Auten JJ, Lowe DK. Ado-trastuzumab emtansine: a HER2-positive targeted antibody-drug conjugate. Ann Pharmacother. 2014;48(11):1484–1493. [DOI] [PubMed] [Google Scholar]
  • 15. Welslau M, Diéras V, Sohn JH et al. Patient-reported outcomes from EMILIA, a randomized phase 3 study of trastuzumab emtansine (T-DM1) versus capecitabine and lapatinib in human epidermal growth factor receptor 2-positive locally advanced or metastatic breast cancer. Cancer. 2014;120(5):642–651. [DOI] [PubMed] [Google Scholar]
  • 16. Bence AK, Anderson EB, Halepota MA et al. Phase I pharmacokinetic studies evaluating single and multiple doses of oral GW572016, a dual EGFR-ErbB2 inhibitor, in healthy subjects. Invest New Drugs. 2005;23(1):39–49. [DOI] [PubMed] [Google Scholar]
  • 17. Amado RG, Wolf M, Peeters M et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26(10):1626–1634. [DOI] [PubMed] [Google Scholar]
  • 18. Maritaz C, Metz C, Baba-Hamed N, Jardin-Szucs M, Deplanque G. Cetuximab-induced aseptic meningitis: case report and review of a rare adverse event. BMC Cancer. 2016;16:384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Paz-Ares L, Mezger J, Ciuleanu TE et al. ; INSPIRE investigators. Necitumumab plus pemetrexed and cisplatin as first-line therapy in patients with stage IV non-squamous non-small-cell lung cancer (INSPIRE): an open-label, randomised, controlled phase 3 study. Lancet Oncol. 2015;16(3):328–337. [DOI] [PubMed] [Google Scholar]
  • 20. Kanaan Z, Kulairi Z, Titianu M, Saha S, Kumar S. Guillain-Barré syndrome following treatment with sunitinib malate. Case Rep Oncol Med. 2014;2014:712040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Mulherin B, Loconte NK, Holen KD. Guillain-Barre syndrome after treatment with sunitinib malate? Oncology. 2008;1:66–67, 70–71. [PubMed] [Google Scholar]
  • 22. Martín G, Bellido L, Cruz JJ. Reversible posterior leukoencephalopathy syndrome induced by sunitinib. J Clin Oncol. 2007;25(23):3559. [DOI] [PubMed] [Google Scholar]
  • 23. Kamiya-Matsuoka C, Cachia D, Olar A, Armstrong TS, Gilbert MR. Primary brain tumors and posterior reversible encephalopathy syndrome. Neurooncol Pract. 2014;1(4):184–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Broniscer A, Baker JN, Tagen M et al. Phase I study of vandetanib during and after radiotherapy in children with diffuse intrinsic pontine glioma. J Clin Oncol. 2010;28(31):4762–4768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Kenney PA, Wood CG. Words of wisdom. Re: Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Eur Urol. 2012;62(1):182–183. [DOI] [PubMed] [Google Scholar]
  • 26. Locati LD, Licitra L, Agate L et al. Treatment of advanced thyroid cancer with axitinib: phase 2 study with pharmacokinetic/pharmacodynamic and quality-of-life assessments. Cancer. 2014;120(17):2694–2703. [DOI] [PubMed] [Google Scholar]
  • 27. Levy A, Benmoussa L, Ammari S, Albiges L, Escudier B. Reversible posterior leukoencephalopathy syndrome induced by axitinib. Clin Genitourin Cancer. 2014;12(1):e33–e34. [DOI] [PubMed] [Google Scholar]
  • 28. Cabanillas ME, Habra MA. Lenvatinib: role in thyroid cancer and other solid tumors. Cancer Treat Rev. 2016;42:47–55. [DOI] [PubMed] [Google Scholar]
  • 29. Demirci NS, Erdem GU, Dogan M, Ozdemir NY, Zengin N. A rare case: hallucination associated with pazopanib. J Cancer Res Ther. 2015;11(4):961–962. [DOI] [PubMed] [Google Scholar]
  • 30. Chelis L, Souftas V, Amarantidis K et al. Reversible posterior leukoencephalopathy syndrome induced by pazopanib. BMC Cancer. 2012;12:489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Advani RH, Buggy JJ, Sharman JP et al. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignancies. J Clin Oncol. 2013;31(1):88–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Seiter K, Stiefel MF, Barrientos J et al. Successful treatment of ibrutinib-associated central nervous system hemorrhage with platelet transfusion support. Stem Cell Investig. 2016;3:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Kamel S, Horton L, Ysebaert L et al. Ibrutinib inhibits collagen-mediated but not ADP-mediated platelet aggregation. Leukemia. 2015;29(4):783–787. [DOI] [PubMed] [Google Scholar]
  • 34. Bernard S, Goldwirt L, Amorim S et al. Activity of ibrutinib in mantle cell lymphoma patients with central nervous system relapse. Blood. 2015;126(14):1695–1698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Dreyling M, Jurczak W, Jerkeman M et al. Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: an international, randomised, open-label, phase 3 study. Lancet. 2016;387(10020):770–778. [DOI] [PubMed] [Google Scholar]
  • 36. Lutz M, Schulze AB, Rebber E et al. Progressive multifocal leukoencephalopathy after ibrutinib therapy for chronic lymphocytic leukemia. Cancer Res Treat. 2017;49(2):548–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Low PC, Manzanero S, Mohannak N et al. PI3Kδ inhibition reduces TNF secretion and neuroinflammation in a mouse cerebral stroke model. Nat Commun. 2014;5:3450. [DOI] [PubMed] [Google Scholar]
  • 38. Chun SG, Iyengar P, Gerber DE, Hogan RN, Timmerman RD. Optic neuropathy and blindness associated with crizotinib for non-small-cell lung cancer with EML4-ALK translocation. J Clin Oncol. 2015;33(5):e25–e26. [DOI] [PubMed] [Google Scholar]
  • 39. Ou SH, Ahn JS, De Petris L et al. Alectinib in crizotinib-refractory ALK-rearranged non-small-cell lung cancer: a phase II global study. J Clin Oncol. 2016;34(7):661–668. [DOI] [PubMed] [Google Scholar]
  • 40. Landi L, Cappuzzo F. Ceritinib for the treatment of patients with anaplastic lymphoma kinase (ALK)-positive metastatic non-small cell lung cancer. Expert Rev Clin Pharmacol. 2016;9(2):203–214. [DOI] [PubMed] [Google Scholar]
  • 41. Tan DS, Araújo A, Zhang J et al. Comparative efficacy of ceritinib and crizotinib as initial ALK-targeted therapies in previously treated advanced NSCLC: an adjusted comparison with external controls. J Thorac Oncol. 2016;11(9):1550–1557. [DOI] [PubMed] [Google Scholar]
  • 42. Shen L, Ji HF. Ceritinib in ALK-rearranged non-small-cell lung cancer. N Engl J Med. 2014;370(26):2537. [DOI] [PubMed] [Google Scholar]
  • 43. Cooper MR, Chim H, Chan H, Durand C. Ceritinib: a new tyrosine kinase inhibitor for non-small-cell lung cancer. Ann Pharmacother. 2015;49(1):107–112. [DOI] [PubMed] [Google Scholar]
  • 44. Katayama R, Kobayashi Y, Friboulet L et al. Cabozantinib overcomes crizotinib resistance in ROS1 fusion-positive cancer. Clin Cancer Res. 2015;21(1):166–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Chong CR, Bahcall M, Capelletti M et al. Identification of existing drugs that effectively target NTRK1 and ROS1 rearrangements in lung cancer. Clin Cancer Res. 2017;23(1):204–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Hecht M, Zimmer L, Loquai C et al. Radiosensitization by BRAF inhibitor therapy-mechanism and frequency of toxicity in melanoma patients. Ann Oncol. 2015;26(6):1238–1244. [DOI] [PubMed] [Google Scholar]
  • 47. Anker CJ, Grossmann KF, Atkins MB, Suneja G, Tarhini AA, Kirkwood JM. Avoiding severe toxicity from combined BRAF inhibitor and radiation treatment: consensus guidelines from the Eastern Cooperative Oncology Group (ECOG). Int J Radiat Oncol Biol Phys. 2016;95(2):632–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Kroeze SG, Fritz C, Hoyer M et al. Toxicity of concurrent stereotactic radiotherapy and targeted therapy or immunotherapy: a systematic review. Cancer Treat Rev. 2017;53:25–37. [DOI] [PubMed] [Google Scholar]
  • 49. Shailesh FN, Singh M, Tiwari U, Hutchins LF. Vemurafenib-induced bilateral facial palsy. J Postgrad Med. 2014;60(2):187–188. [DOI] [PubMed] [Google Scholar]
  • 50. Klein O, Ribas A, Chmielowski B et al. Facial palsy as a side effect of vemurafenib treatment in patients with metastatic melanoma. J Clin Oncol. 2013;31(12):e215–e217. [DOI] [PubMed] [Google Scholar]
  • 51. Flaherty KT, Infante JR, Daud A et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 2012;367(18):1694–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Schadendorf D, Little S, Rubin S et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med. 2015;372(1):30–39. [DOI] [PubMed] [Google Scholar]
  • 53. DeMichele A, Clark AS, Tan KS et al. CDK 4/6 inhibitor palbociclib (PD0332991) in Rb+ advanced breast cancer: phase II activity, safety, and predictive biomarker assessment. Clin Cancer Res. 2015;21(5):1001–995. [DOI] [PubMed] [Google Scholar]
  • 54. Ramelyte E, Amann VC, Dummer R. Sonidegib for the treatment of advanced basal cell carcinoma. Expert Opin Pharmacother. 2016;17(14):1963–1968. [DOI] [PubMed] [Google Scholar]
  • 55. Macdonald JB, Macdonald B, Golitz LE, LoRusso P, Sekulic A. Cutaneous adverse effects of targeted therapies: part I: inhibitors of the cellular membrane. J Am Acad Dermatol. 2015;72(2):203–218; quiz 219. [DOI] [PubMed] [Google Scholar]
  • 56. Subramanian S, Bates SE, Wright JJ, Espinoza-Delgado I, Piekarz RL. Clinical toxicities of histone deacetylase inhibitors. Pharmaceuticals (Basel). 2010;3(9):2751–2767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Qin W, Tan CY, Huang X, Huang Z, Tao Y, Fu P. Rapamycin-induced posterior reversible encephalopathy in a kidney transplantation patient. Int Urol Nephrol. 2011;43(3):913–916. [DOI] [PubMed] [Google Scholar]
  • 58. Pallet N, Legendre C. Adverse events associated with mTOR inhibitors. Expert Opin Drug Saf. 2013;12(2):177–186. [DOI] [PubMed] [Google Scholar]
  • 59. Shaw HM, Hall M. Emerging treatment options for recurrent ovarian cancer: the potential role of olaparib. Onco Targets Ther. 2013;6:1197–1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Swisher EM, Lin KK, Oza AM et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol. 2017;18(1):75–87. [DOI] [PubMed] [Google Scholar]
  • 61. Cortes J, Lipton JH, Rea D et al. ; Omacetaxine 202 Study Group. Phase 2 study of subcutaneous omacetaxine mepesuccinate after TKI failure in patients with chronic-phase CML with T315I mutation. Blood. 2012;120(13):2573–2580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Moreau P, Masszi T, Grzasko N et al. ; TOURMALINE-MM1 Study Group. Oral ixazomib, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med. 2016;374(17):1621–1634. [DOI] [PubMed] [Google Scholar]
  • 63. Steele JM. Carfilzomib: a new proteasome inhibitor for relapsed or refractory multiple myeloma. J Oncol Pharm Pract. 2013;19(4):348–354. [DOI] [PubMed] [Google Scholar]
  • 64. Muz B, Ghazarian RN, Ou M, Luderer MJ, Kusdono HD, Azab AK. Spotlight on ixazomib: potential in the treatment of multiple myeloma. Drug Des Devel Ther. 2016;10:217–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Schlafer D, Shah KS, Panjic EH, Lonial S. Safety of proteasome inhibitors for treatment of multiple myeloma. Expert Opin Drug Saf. 2016:1–17. [DOI] [PubMed] [Google Scholar]
  • 66. Roberts AW, Davids MS, Pagel JM et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016;374(4):311–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Hill BT, Kalaycio M. Profile of obinutuzumab for the treatment of patients with previously untreated chronic lymphocytic leukemia. Onco Targets Ther. 2015;8:2391–2397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Raisch DW, Rafi JA, Chen C, Bennett CL. Detection of cases of progressive multifocal leukoencephalopathy associated with new biologicals and targeted cancer therapies from the FDA’s adverse event reporting system. Expert Opin Drug Saf. 2016;15(8):1003–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Berger JR, Aksamit AJ, Clifford DB et al. PML diagnostic criteria: consensus statement from the AAN Neuroinfectious Disease Section. Neurology. 2013;80(15):1430–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Mukherjee S, Chatterjee G, Ghosh M, Das B, Majumder D. Efficacy and toxicity assessment of different antibody based antiangiogenic drugs by computational docking method. Adv Bioinformatics. 2016;2016:7053712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Tlemsani C, Mir O, Boudou-Rouquette P et al. Posterior reversible encephalopathy syndrome induced by anti-VEGF agents. Target Oncol. 2011;6(4):253–258. [DOI] [PubMed] [Google Scholar]
  • 72. Seet RC, Rabinstein AA. Clinical features and outcomes of posterior reversible encephalopathy syndrome following bevacizumab treatment. QJM. 2012;105(1):69–75. [DOI] [PubMed] [Google Scholar]
  • 73. Van Cutsem E, Tabernero J, Lakomy R et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol. 2012;30(28):3499–3506. [DOI] [PubMed] [Google Scholar]
  • 74. Young K, Smyth E, Chau I. Ramucirumab for advanced gastric cancer or gastro-oesophageal junction adenocarcinoma. Therap Adv Gastroenterol. 2015;8(6):373–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Tap WD, Jones RL, Van Tine BA et al. Olaratumab and doxorubicin versus doxorubicin alone for treatment of soft-tissue sarcoma: an open-label phase 1b and randomised phase 2 trial. Lancet. 2016;388(10043):488–497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Ploessl C, Pan A, Maples KT, Lowe DK. Dinutuximab: an anti-GD2 monoclonal antibody for high-risk neuroblastoma. Ann Pharmacother. 2016;50(5):416–422. [DOI] [PubMed] [Google Scholar]
  • 77. Lonial S, Weiss BM, Usmani SZ et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet. 2016;387(10027):1551–1560. [DOI] [PubMed] [Google Scholar]
  • 78. Lonial S, Dimopoulos M, Palumbo A et al. ; ELOQUENT-2 Investigators. Elotuzumab therapy for relapsed or refractory multiple myeloma. N Engl J Med. 2015;373(7):621–631. [DOI] [PubMed] [Google Scholar]
  • 79. Nagorsen D, Baeuerle PA. Immunomodulatory therapy of cancer with T cell-engaging BiTE antibody blinatumomab. Exp Cell Res. 2011;317(9):1255–1260. [DOI] [PubMed] [Google Scholar]
  • 80. Topp MS, Gökbuget N, Stein AS et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 2015;16(1):57–66. [DOI] [PubMed] [Google Scholar]
  • 81. Topp MS, Gökbuget N, Zugmaier G et al. Phase II trial of the anti-CD19 bispecific T cell-engager blinatumomab shows hematologic and molecular remissions in patients with relapsed or refractory B-precursor acute lymphoblastic leukemia. J Clin Oncol. 2014;32(36):4134–4140. [DOI] [PubMed] [Google Scholar]
  • 82. Folan SA, Rexwinkle A, Autry J, Bryan JC. Blinatumomab: bridging the gap in adult relapsed/refractory B-cell acute lymphoblastic leukemia. Clin Lymphoma Myeloma Leuk. 2016;16(Suppl):S2–S5. [DOI] [PubMed] [Google Scholar]
  • 83. Nagorsen D, Kufer P, Baeuerle PA, Bargou R. Blinatumomab: a historical perspective. Pharmacol Ther. 2012;136(3):334–342. [DOI] [PubMed] [Google Scholar]
  • 84. Viardot A, Goebeler ME, Hess G et al. Phase 2 study of the bispecific T-cell engager (BiTE) antibody blinatumomab in relapsed/refractory diffuse large B-cell lymphoma. Blood. 2016;127(11):1410–1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Lejeune FJ. The conquest of melanoma by immunotherapy. Melanoma Res. 2015;25(5):373–375. [DOI] [PubMed] [Google Scholar]
  • 86. Bertrand A, Kostine M, Barnetche T, Truchetet ME, Schaeverbeke T. Immune related adverse events associated with anti-CTLA-4 antibodies: systematic review and meta-analysis. BMC Med. 2015;13:211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Horvat TZ, Adel NG, Dang TO et al. Immune-related adverse events, need for systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with ipilimumab at Memorial Sloan Kettering Cancer Center. J Clin Oncol. 2015;33(28):3193–3198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Cao Y, Nylander A, Ramanan S et al. CNS demyelination and enhanced myelin-reactive responses after ipilimumab treatment. Neurology. 2016;86(16):1553–1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Gettings EJ, Hackett CT, Scott TF. Severe relapse in a multiple sclerosis patient associated with ipilimumab treatment of melanoma. Mult Scler. 2015;21(5):670. [DOI] [PubMed] [Google Scholar]
  • 90. Maurice C, Schneider R, Kiehl TR et al. Subacute CNS demyelination after treatment with nivolumab for melanoma. Cancer Immunol Res. 2015;3(12):1299–1302. [DOI] [PubMed] [Google Scholar]
  • 91. Liao B, Shroff S, Kamiya-Matsuoka C, Tummala S. Atypical neurological complications of ipilimumab therapy in patients with metastatic melanoma. Neuro Oncol. 2014;16(4):589–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Thaipisuttikul I, Chapman P, Avila EK. Peripheral neuropathy associated with ipilimumab: a report of 2 cases. J Immunother. 2015;38(2):77–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Zimmer L, Goldinger SM, Hofmann L et al. Neurological, respiratory, musculoskeletal, cardiac and ocular side-effects of anti-PD-1 therapy. Eur J Cancer. 2016;60:210–225. [DOI] [PubMed] [Google Scholar]
  • 94. de Maleissye MF, Nicolas G, Saiag P. Pembrolizumab-induced demyelinating polyradiculoneuropathy. N Engl J Med. 2016;375(3):296–297. [DOI] [PubMed] [Google Scholar]
  • 95. Gonzalez NL, Puwanant A, Lu A, Marks SM, Živković SA. Myasthenia triggered by immune checkpoint inhibitors: new case and literature review. Neuromuscul Disord. 2017;27(3):266–268. [DOI] [PubMed] [Google Scholar]
  • 96. Sheik Ali S, Goddard AL, Luke JJ et al. Drug-associated dermatomyositis following ipilimumab therapy: a novel immune-mediated adverse event associated with cytotoxic T-lymphocyte antigen 4 blockade. JAMA Dermatol. 2015;151(2):195–199. [DOI] [PubMed] [Google Scholar]
  • 97. Valecha GK, Vennepureddy A, Ibrahim U, Safa F, Samra B, Atallah JP. Anti-PD-1/PD-L1 antibodies in non-small cell lung cancer: the era of immunotherapy. Expert Rev Anticancer Ther. 2017;17(1):47–59. [DOI] [PubMed] [Google Scholar]
  • 98. Hottinger AF. Neurologic complications of immune checkpoint inhibitors. Curr Opin Neurol. 2016;29(6):806–812. [DOI] [PubMed] [Google Scholar]
  • 99. Harrison TS, Lyseng-Williamson KA. Vincristine sulfate liposome injection: a guide to its use in refractory or relapsed acute lymphoblastic leukemia. BioDrugs. 2013;27(1):69–74. [DOI] [PubMed] [Google Scholar]
  • 100. Hagemeister F, Rodriguez MA, Deitcher SR et al. Long term results of a phase 2 study of vincristine sulfate liposome injection (Marqibo®) substituted for non-liposomal vincristine in cyclophosphamide, doxorubicin, vincristine, prednisone with or without rituximab for patients with untreated aggressive non-Hodgkin lymphomas. Br J Haematol. 2013;162(5):631–638. [DOI] [PubMed] [Google Scholar]
  • 101. O’Brien S, Schiller G, Lister J et al. High-dose vincristine sulfate liposome injection for advanced, relapsed, and refractory adult Philadelphia chromosome-negative acute lymphoblastic leukemia. J Clin Oncol. 2013;31(6):676–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Wozniak KM, Vornov JJ, Wu Y et al. Sustained accumulation of microtubule-binding chemotherapy drugs in the peripheral nervous system: correlations with time course and neurotoxic severity. Cancer Res. 2016;76(11):3332–3339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Noguchi Y, Kawashima Y, Kawara H, Kaneko M, Nakauchi H, Tokuyama Y. An undeniable case of optic neuropathy due to cabazitaxel. Gan To Kagaku Ryoho. 2016;43(6):777–779. [PubMed] [Google Scholar]
  • 104. Omlin A, Sartor O, Rothermundt C et al. Analysis of side effect profile of alopecia, nail changes, peripheral neuropathy, and dysgeusia in prostate cancer patients treated with docetaxel and cabazitaxel. Clin Genitourin Cancer. 2015;13(4):e205–e208. [DOI] [PubMed] [Google Scholar]
  • 105. van de Donk N, Dhimolea E. Brentuximab vedotin. MAbs. 2012;4(4):458–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Chen R, Gopal AK, Smith SE et al. Five-year survival and durability results of brentuximab vedotin in patients with relapsed or refractory Hodgkin lymphoma. Blood. 2016;128(12):1562–1566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Ho AL, Lipson BL, Sherman EJ et al. A phase II study of pralatrexate with vitamin B12 and folic acid supplementation for previously treated recurrent and/or metastatic head and neck squamous cell cancer. Invest New Drugs. 2014;32(3):549–554. [DOI] [PubMed] [Google Scholar]
  • 108. Burness CB, Duggan ST. Trifluridine/tipiracil: a review in metastatic colorectal cancer. Drugs. 2016;76(14):1393–1402. [DOI] [PubMed] [Google Scholar]
  • 109. Desar IM, Constantinidou A, Kaal SE, Jones RL, van der Graaf WT. Advanced soft-tissue sarcoma and treatment options: critical appraisal of trabectedin. Cancer Manag Res. 2016;8:95–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Le Cesne A, Blay JY, Domont J et al. Interruption versus continuation of trabectedin in patients with soft-tissue sarcoma (T-DIS): a randomised phase 2 trial. Lancet Oncol. 2015;16(3):312–319. [DOI] [PubMed] [Google Scholar]
  • 111. Patel UH, Mir MA, Sivik JK, Raheja D, Pandey MK, Talamo G. Central neurotoxicity of immunomodulatory drugs in multiple myeloma. Hematol Rep. 2015;7(1):5704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Dalla Torre C, Zambello R, Cacciavillani M et al. Lenalidomide long-term neurotoxicity: clinical and neurophysiologic prospective study. Neurology. 2016;87(11):1161–1166. [DOI] [PubMed] [Google Scholar]
  • 113. Offidani M, Corvatta L, Caraffa P et al. Pomalidomide for the treatment of relapsed-refractory multiple myeloma: a review of biological and clinical data. Expert Rev Anticancer Ther. 2014;14(5):499–510. [DOI] [PubMed] [Google Scholar]
  • 114. Fizazi K, Scher HI, Molina A et al. ; COU-AA-301 Investigators. Abiraterone acetate for treatment of metastatic castration-resistant prostate cancer: final overall survival analysis of the COU-AA-301 randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2012;13(10):983–992. [DOI] [PubMed] [Google Scholar]
  • 115. Scher HI, Fizazi K, Saad F et al. ; AFFIRM Investigators. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367(13):1187–1197. [DOI] [PubMed] [Google Scholar]
  • 116. Rick FG, Block NL, Schally AV. An update on the use of degarelix in the treatment of advanced hormone-dependent prostate cancer. Onco Targets Ther. 2013;6:391–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Kantoff PW, Higano CS, Shore ND et al. ; IMPACT Study Investigators. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411–422. [DOI] [PubMed] [Google Scholar]
  • 118. Andtbacka RH, Kaufman HL, Collichio F et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 2015;33(25):2780–2788. [DOI] [PubMed] [Google Scholar]
  • 119. Wolach O, Stone RM. Blinatumomab for the treatment of Philadelphia chromosome-negative, precursor b-cell acute lymphoblastic leukemia. Clin Cancer Res. 2015;21(19):4262–4269. [DOI] [PubMed] [Google Scholar]
  • 120. Jena B, Dotti G, Cooper LJ. Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood. 2010;116(7):1035–1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Grupp SA, Kalos M, Barrett D et al. Chimeric antigen receptor–modified T cells in chronic lymphoid leukemia. N Engl J Med. 2016;374(10):998–998. [DOI] [PubMed] [Google Scholar]
  • 122. Dai H, Zhang W, Li X et al. Tolerance and efficacy of autologous or donor-derived T cells expressing CD19 chimeric antigen receptors in adult B-ALL with extramedullary leukemia. Oncoimmunology. 2015;4(11):e1027469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Lee DW, Kochenderfer JN, Stetler-Stevenson M et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015;385(9967):517–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Tanaka T, Narazaki M, Kishimoto T. Immunotherapeutic implications of IL-6 blockade for cytokine storm. Immunotherapy. 2016;8(8):959–970. [DOI] [PubMed] [Google Scholar]

Articles from Neuro-Oncology are provided here courtesy of Society for Neuro-Oncology and Oxford University Press

RESOURCES