Abstract
Platinum-induced peripheral neurotoxicity (PIPN) is a common side effect of platinum-based chemotherapy that may cause dose reduction and discontinuation, with oxaliplatin being more neurotoxic. PIPN includes acute neurotoxicity restricted to oxaliplatin, and chronic non-length dependent sensory neuronopathy with positive and negative sensory symptoms and neuropathic pain in both upper and lower limbs. Chronic sensory axonal neuropathy manifesting as stocking-and-glove distribution is also frequent. Worsening of neuropathic symptoms after completing the last chemotherapy course may occur. Motor and autonomic involvement is uncommon. Ototoxicity is frequent in children and more commonly to cisplatin. Platinum-based compounds result in more prolonged neuropathic symptoms in comparison to other chemotherapy agents.
Patient reported outcomes questionnaires, clinical evaluation and instrumental tools offer complementary information in PIPN. Electrodiagnostic features include diffusely reduced/abolished sensory action potentials, in keeping with a sensory neuronopathy. PIPN is dependent on cumulative dose but there is a large variability in its occurrence. The search for additional risk factors for PIPN has thus far yielded no consistent findings. There are currently no neuroprotective strategies to reduce the risk of PIPN, and symptomatic treatment is limited to duloxetine that was found effective in a single phase III intervention study. This review critically examines the pathogenesis, incidence, risk factors (both clinical and pharmacogenetic), clinical phenotype and management of PIPN.
Keywords: chemotherapy, neurotoxicity, neuropathy, carboplatin, cisplatin, oxaliplatin, assessment, diagnosis, prevention, treatment
Introduction
Platinum-based chemotherapy agents have been used for cancer treatment ever since the introduction of cisplatin in the 1970s, which followed the discovery of its anti-proliferative properties by Rosenberg and colleagues1, 2. Subsequently, the newer platinum derivatives, carboplatin and oxaliplatin, have been incorporated widely into the oncologist’s armamentarium3. Unfortunately, despite their efficacy against a variety of solid tumors, the platinum compounds have several serious side effects that often limit their use. While some of the platinum-based chemotherapeutic side effects (myelosuppression, nephrotoxicity) may be mitigated, neurotoxicity continues to be a serious dose-limiting side effect that is without any proven preventative strategy. In this review, we aim to critically examine the clinical features of peripheral neurotoxicity due to platinum-based chemotherapy, discuss putative risk factors for its development, and describe clinical trials aimed at reducing its impact on patients with cancer.
Search strategy and selection criteria
We performed a systematic search in PubMed and Scopus for relevant original research papers written in English reporting on humans from 1990 up to date with terms: ‘chemotherapy-induced peripheral neurotoxicity’, ‘toxic neuropathy’, ‘peripheral neuropathy’, ‘neurotoxicity syndromes’ combined with ‘cisplatin’, ‘oxaliplatin’, ‘carboplatin’. We focused on controlled studies and large case series with adequate sample size, but selected single case reports or small case series were included for less established topics.
Pathogenesis of Platinum-induced Peripheral Neurotoxicity
The sensory neurons of the dorsal root ganglia (DRG) are the primary target of peripheral neurotoxicity arising from platinum-based chemotherapy. It is widely accepted that binding of platinum to DNA is the primary inciting mechanism that leads to chronic damage to the DRG neuron, which can ultimately trigger apoptosis due to aberrant re-entry into the cell cycle pathway4, 5. Apoptosis of DRG neurons, as a result of mitochondrial dysfunction, oxidative stress and formation of DNA adducts and cross-links, leads to a non-length-dependent sensory neuronopathy (or ganglionopathy), which is a clinically-distinct syndrome from other causes of chemotherapy-induced peripheral neuropathy (CIPN) that tend to be a length-dependent or stocking-and-glove pattern. There is further evidence that there is not only damage to nuclear DNA, but also significant binding and damage to mitochondrial DNA6, which may account for longer-term aspects of neuronal damage in patients with platinum-induced peripheral neurotoxicity (PIPN), such as ‘coasting’ and length-dependent sensory neuropathic features.
Oxaliplatin is unique in that it also produces an acute neurotoxicity, characterized by cold-induced dysesthesias. The acute oxaliplatin neurotoxicity appears to be mediated by increased neuronal excitability and altered axonal refractoriness, possibly via oxaliplatin interaction with voltage-gated sodium channels and changes in kinetics of sodium channel inactivation7. Additionally, there appears to be an association between development of acute oxaliplatin neurotoxicity and susceptibility to the chronic neuropathy8, which is intriguing given the disparate mechanisms of toxicity.
Clinical Aspects of Platinum-induced Peripheral Neurotoxicity
Clinical Presentations
PIPN includes acute and chronic peripheral neurotoxicity, the latter associated to all platinum-based compounds, while the former is restricted to oxaliplatin. Furthermore, it is generally regarded that cisplatin is more ototoxic, carboplatin is more myelosuppressive, and oxaliplatin is more neurotoxic 9. A prospective, multicenter study focusing on oxaliplatin-induced peripheral neurotoxicity showed the acute and chronic neurotoxicity to occur in 85% and 73% of patients, respectively 10.
In keeping with the view that platinum-based compounds exert their toxicity at the sensory neuronal cell body, the chronic peripheral neurotoxicity primarily presents as a sensory neuronopathy (or ganglionopathy) with anterograde non-length dependent neuronal degeneration 11. Positive sensory symptoms (numbness, tingling, paresthesia, neuropathic pain) and loss-of-function signs (reduction/loss of sensation to touch, vibration and joint position with reduced/absent deep tendon reflexes) usually appear in both upper and lower limbs. With prolonged treatment the neuronopathy can eventually evolve to other body segments in a non-length dependent pattern with sensory ataxia12. Additionally, chronic neuropathy manifesting as a pure sensory, stocking-and-glove distribution, axonal neuropathy has been observed in 50–70% of patients 13-16. A long-term study showed sensory symptoms to be more prominent in the hands than the feet during chemotherapy; whereas by 18 months, symptoms were more severe in the feet than the hands 8. Finally, Lhermitte’s phenomenon has been reported in some patients, presumably secondary to DRG axonal degeneration within the spinal cord17.
Motor and autonomic symptoms and signs are uncommon in patients treated with platinum-based compounds, but may be observed in severe cases 18. Therefore, when motor and autonomic symptoms are predominant and/or there is an absence of sensory involvement, an alternative diagnosis should be considered. As an example, platinum-based compounds have been rarely associated with acute inflammatory demyelinating polyradiculoneuritis in patients with solid tumor 19-23, which appears to respond well to standard immunotherapy. Additionally, it is important to be aware that treatment with more than one neurotoxic agent may increase the likelihood of peripheral neuropathy24, which may be associated with involvement of the autonomic system (especially parasympathetic heart innervation) 25.
Hearing loss and tinnitus are a progressive and irreversible adverse effect of platinum-based chemotherapy, frequently reported bilaterally in children and more commonly occurring to cisplatin than oxaliplatin 26. Approximately 60% (range 26–90%) children receiving cisplatin develop irreversible bilateral hearing loss27-31, which may have a deleterious effect in their speech, language, and social skills 27. Cisplatin exposure has also been associated with cognitive dysfunction, the so-called “chemo-brain”, which is beyond the scope of this review 32.
Time course
With all platinum-based compounds, chronic sensory neuropathy/neuronopathy development occurs with increasing cumulative dosage. As already stated, oxaliplatin is unique as it typically also causes acute neurotoxicity. The oxaliplatin acute neurotoxicity is characterized by transient paresthesia, dysesthesia and muscle cramps induced by cold exposure, a phenomenon often called cold allodynia or hyperalgesia that typically resolves before the next oxaliplatin cycle 33. Symptoms reported by patients include paresthesia and/or dysesthesia in the perioral, pharyngeal and laryngeal regions, as well as in the distal limbs triggered and exacerbated by cold (especially when drinking beverages), abnormal breathing and swallowing, muscle spasms or cramps among other uncommon symptoms related to the acute oxaliplatin hyperexcitability syndrome34, 35. Acute peripheral neurotoxicity to oxaliplatin, documented either by clinical symptoms or quantitative sensory testing, has been demonstrated to be associated with the development of chronic peripheral neuropathy. This observation has the potential of making the acute neurotoxicity a predictor for selecting patients who may benefit from neuroprotective strategies 35, 36.
The large majority (89%) of a cohort of 346 patients under fluorouracil, leucovorin, and oxaliplatin (FOLFOX) reported at least one symptom of acute oxaliplatin neurotoxicity within the first cycle, namely sensitivity to cold (71%), throat discomfort (63%), or muscle cramps (42%), with a peak at day 3, later improvement, and incomplete remission between treatments 8. According to a systematic review, acute oxaliplatin neuropathy (Common Terminology Criteria Adverse Events, CTCAE grades 1–4) occurred in 4–98% of patients, with moderate to severe toxicities commonly encountered in patients receiving a large dose of oxaliplatin (> 85 mg/m2) and/ or combined drugs 37.
Another important feature of PIPN is ‘coasting’, i.e. worsening of neuropathy signs and symptoms for months after completing the last chemotherapy course8. This is an important factor in deciding when to discontinue therapy due to neuropathic symptoms, as impairments and disability may progress once the drug has been stopped. It has been hypothesized that coasting may be due to drug accumulation in the DRG and/or chronic neuronal damage from mitochondrial platinum-DNA adducts that impair mitochondrial function6.
Incidence and range of severity
The incidence of chronic PIPN is related to cumulative dose and dose per cycle 38, which in the case of cisplatin usually occurs with administration of 250–450 mg/m2 38, 39. Almost all patients develop neuropathy after a cumulative cisplatin dose of 500–600 mg/ m2, which is severely disabling in approximately 10% of patients 12. Studies investigating the association between chronic peripheral neurotoxicity and oxaliplatin doses reported threshold cumulative dose in symptomatic patients to range from 850 to 1800 mg/m2 40-43. Carboplatin-induced peripheral neurotoxicity is less frequent and severe at conventional doses 44.
Incidence and severity of PIPN vary across studies12. Using a variety of neurological outcome measures, Phase III and prospective studies have reported cisplatin-induced peripheral neurotoxicity to occur as Grade 1 in 14–33%, Grade 2 in 0–33%, Grade 3 in 2–19% and Grade 4 in 0–4% of patients (Table 1). Likewise, oxaliplatin was reported to cause peripheral neurotoxicity of Grade 1 in 21–94%, Grade 2 in 5–42%, and Grade 3–4 in 3–19% 12 (Table 2). When neuropathy was assessed with specific neurological assessment tools (unlike the CTCAE), peripheral neuropathy was found in 17–100% of patients for cisplatin and 50–75% for oxaliplatin12. These findings suggest that, apart from the cumulative dose and dose per cycle that are known to influence the risk of PIPN, the ascertainment/diagnostic criteria strongly influence epidemiological data. Because of the heterogeneous tools utilized in the studies, a formal framework for the definition, classification and measurements of PIPN is of great importance 45.
Table 1.
Study | Design | Pts (N) |
Cancer type(s) | Clinical features of cisplatin-induced peripheral neuropathy | ||
---|---|---|---|---|---|---|
Assessment methods | Duration of follow-up | Incidence | ||||
Swenerton (1992)116 | Phase III | 210 | Ovarian | ECOG | After 6 CMT cycles | Grades 1-2: 52%; grade 3: 1% |
Kemp (1996)117 | Neuroprotection | 120 | Ovarian | NCI-CTC | Up to CMT cycle 6 | Grades 1-2: 55%; grade 3: 13% |
Wozniak (1998)118 | Phase III | 200 | NSCLC | SWOG | 1 year after CMT completion | Grades 3-4: 1% |
Sutton (2000)119 | Phase III | 90 | Uterus carcinosarcoma | GOG | Up to CMT cycle 8 | Grades 2: 8%; grade 3-4: 13% |
Gatzmeier (2000)120 | Phase III | 206 | NSCLC | NCI-CTC, QLQ-C30 | Up to median 3 months after CMT | Grades 1-2: 12%; grades 3-4: 1% |
Wachters (2003)121 | Phase III | 119 | NSCLC | NCI-CTC, QLQ-C30 | 6 months after CMT completion | Grades 1-2: 22%; grades 3-4: 1% |
Fleming (2004)122 | Phase III | 129 | Endometrium | NCI-CTC, FACT/GOG-Ntx | 12 months after CMT completion | Grades 1-2: 3%; grades 3-4: 1% |
Al-Batran (2008)123 | Phase III | 112 | GE | NCI-CTC | Median follow-up 6 months | Grades 1-2: 20%; grades 3-4: 2% |
Homesley (2009)124 | Phase III | 261 | Endometrium | NCI-CTC, FACT/GOG-Ntx | 47 months median follow-up after CMT completion | Grades 1-2: 28%; grades 3-4: 2% |
Chen (2013)125 | Phase III/IVb | 158 | Nasopharingeal | WHO | Median follow-up 70 months | Grade 3-5: 2% |
Friedrich (2013)126 | Prospective | 49 | Medulloblastoma | NCI-CTCAE | Median follow-up 3.7 years | Grades 2-3: 51%, any grade: 74% |
Conroy (2014)127 | Phase II/III | 128 | Oesophagus | NCI-CTCAE | Median follow-up 25.3 months | Grades 1-2: 1%; grades 3-4: 0% |
Yamada (2015)128 | Phase III | 337 | Gastric | NCI-CTCAE | Median follow-up 25.9 months | Grades 3-4: 0%, any grade: 24% |
Bando (2016)129 | Phase III | 343 | Gastric | NCI-CTCAE | Median follow-up 13.5 months | Grades 3-4: 0% |
Fung (2017)130 | Prospective | 952 | Testis | Ad-hoc PRO questionnaire | Median 4.3 years after CMT | Any neuropathy: 21-29% |
Ramaswamy (2017)131 | Prospective | 163 | Gallbladder | NR | NR | Grades 2-3: 3%, |
CMT: chemotherapy; ECOG: Eastern Cooperative Oncology Group criteria; FACT/GOG-Ntx: Functional Assessment of Cancer Therapy Scale/Gynecologic Oncology Group-Neurotoxicity scale; GE: gastroesophageal; GOG: GOG: Gynecologic Oncology Group criteria; NCI-CTC, National Cancer Institute Common Toxicity Criteria; NCI-CTCAE: National Cancer Institute Common Terminology Criteria for Adverse Events; NR: not reported; NSCLC: non-small cell lung cancer; QLQ-C30: European Organization for the Treatment of Cancer quality of life questionnaire-30 items; PRO: Patient Reported Outcome; RCT: Randomized Controlled Trial; SWOG: South West Oncology Group criteria.
Table 2.
Study | Design | Pts (N) |
Cancer type(s) | Clinical features of oxaliplatin-induced peripheral neuropathy | ||
---|---|---|---|---|---|---|
Assessment methods | Duration of follow-up | Incidence | ||||
Giacchetti (2000)132 | Phase III | 100 | CRC | Specific neuropathy scale | Median follow-up 19 months | Grades 1-2: 45%; grades 3-4: 10% |
de Gramont (2000)133 | Phase III | 209 | CRC | NCI-CTC | Median follow-up 8 months | Grades 1-2: 50%; grades 3-4: 18% |
Rothenberg (2003)134 | Phase III | 303 | CRC | NCI-CTC-derived scale | Maximum follow-up 14 months | Grades 1-2: 48-49%; grades 3-4: 2.−3% |
André (2004)135 | Phase III | 1108 | CRC | NCI-CTC | Median follow-up 38 months | Grades 1-2: 80%; grade 3: 12% |
Tournigand (2006)105 | Phase III | 612 | CRC | NCI-CTC | Median follow-up 31 months | Grades 1-2: 71-78%; grades 3-4: 13-18% |
Land (2007)136 | Phase III | 189 | CRC | NCI-CTC, FACT/GOG-Ntx, NCI-Sanofi | Up to 18 months after CMT completion | Grades 1-2: 61-68%; grades 3-4: 3% |
Al-Batran (2008)123 | Phase III | 112 | GE | NCI-CTC, oxaliplatin scale | Median follow-up 6 months | Grades 1-2: 49%; grades 3-4: 14% |
Cassidy (2008)137 | Phase III | 1304 | CRC | NCI-CTC | Median follow-up 30 months | Grades 1-2: 16%; grades 3-4: 4% |
Cunningham (2008)138 | Phase III | 452 | GE | NCI-CTC | Median follow-up 17 months | Grades 1-2: 75%; grades 3-4: 7% |
Allegra (2009)139 | Phase III | 2647 | CRC | NCI-CTC | Up to 1 year after CMT completion | Grade 2: 29-33%; grade 3: 14-16% |
Poplin (2009)140 | Phase III | 272 | Pancreas | NCI-CTC | NR | Grade 3: 10% |
Qvortrup (2010)141 | Phase III | 139 | CRC | NCI-CTC | Median follow-up 14 months | Grade 2: 27-42%; grade 3: 16-19% |
Argyriou (2012)142 | Prospective | 150 | CRC | NCI-CTC, NCS, TNS | Up to 1 month after CMT completion | Grades 1-2: 45-73%; grade 3: 10-18% |
Conroy (2014)127 | Phase II/III | 131 | Oesophagus | NCI-CTCAE | Median follow-up 25.3 months | Grades 1-2: 47%; grades 3-4: 0% |
Yamada (2015)128 | Phase III | 339 | Gastric | NCI-CTCAE | Median follow-up 25.9 months | Grades 3-4: 5%, any grade: 86% |
Bando (2016)129 | Phase III | 343 | Gastric | NCI-CTCAE | Median follow-up 13.5 months | Grades 3-4: 4.5-5.3% |
Lonardi (2016)143 | Phase III | 3759 | CRC | NCI-CTC | Maximum follow-up 3 years | Grade 2: 8-23%; grade 3: 1-8% |
Ramaswamy (2017)131 | Prospective | 163 | Gallbladder | NR | NR | Grades 2-3: 9%, |
Iveson (2018)144 | Phase III | 6022 | CRC | FACT/GOG-Ntx | Maximum follow-up 8 years | Grades 1-2: 77-87%; grades 3-4: 16-18% |
CMT: chemotherapy; CRC: colorectal cancer; FACT/GOG-Ntx: Functional Assessment of Cancer Therapy Scale/Gynecologic Oncology Group-Neurotoxicity scale; GE: gastroesophageal; NCI-CTC, National Cancer Institute Common Toxicity Criteria; NCI-CTCAE: National Cancer Institute Common Terminology Criteria for Adverse Events; NCI-Sanofi: oxaliplatin Sanofi questionnaire; NCS: Nerve Conduction Study; NR: not reported; TNS: Total Neuropathy Score.
Recovery and late effects
When compared to other neurotoxic chemotherapeutics, platinum-based compounds result in more prolonged neuropathic symptoms, presumably due to the presence of irreversible damage to the DRG neuron. Earlier reports suggested chronic PIPN to be reversible after chemotherapy discontinuation 46, but more recent long-term clinical and nerve conduction studies (NCS) demonstrated its persistence up to 2–3 years with complete reversal occurring only in a minority of cases 47, 48. Oxaliplatin also appears to cause more late chronic neurotoxicity when compared to cisplatin. Oxaliplatin-induced peripheral neurotoxicity may be present in 26–46% of patients at 12-month, in 24% of patients at 15–18-month 45, or even in 84% of patients at the 24-month follow-up 47. On the other hand, cisplatin induced peripheral neurotoxicity may persist in 5–20% patients at one year 49, 50.
Interestingly, the chronic form of PIPN is reported to show less of a neuronopathy clinical picture, whereas there are increased length-dependent stocking-and-glove sensory polyneuropathy signs and symptoms 13-16. Furthermore, the degree of intraepidermal nerve fiber loss in distal and proximal biopsies failed to demonstrate a neuronopathy pattern in studies of oxaliplatin-induced neuropathy51, 52. These findings suggest the presence of reversible neurotoxic effects and/or a length-dependent neuropathic component that arises with time. A cross-sectional observational study involving 121 childhood cancer survivor patients showed persistence of neuropathic symptoms in 53% patients after a median of 8.5 years53. In this study, late effects were primarily lower limb predominant sensory dysesthesia plus functional deficits of manual dexterity, distal sensation, and balance53.
Electrophysiology and Imaging
Traditional NCS often demonstrate a sensory neuronopathy pattern in PIPN due to involvement of DRG neurons47, 54, 55. Characteristically, there are diffusely low or absent sensory nerve action potentials (SNAP) with normal sensory conduction velocities, which is not length-dependent. Unless the neurotoxicity is very severe, motor nerves are usually spared. Evidence of primary demyelination is lacking whereas secondary demyelination as a result of severe axonal loss may occur. SNAP abnormalities often precede sensory symptoms 56 and sural SNAP may provide important information in terms of neuropathy progression and has prognostic implications57. Specifically, the dorsal sural nerve (the distal lateral branch of the sural nerve) SNAP has been found to be more sensitive than the sural SNAP for detecting early CIPN 58. In one study, decreased dorsal sural SNAP amplitude, assessed at mid-treatment, was able to predict the neurological outcome at chemotherapy completion 57.
Electrophysiological studies of acute oxaliplatin peripheral neurotoxicity have delineated a primary abnormality in neuronal excitability that may produce the cold-induced dysesthesias. Axonal excitability techniques59 suggest abnormal functioning of axonal voltage-gated Na+ channels in motor and sensory axons with ‘neuromyotonic-type’ discharges in motor axons7, 60, 61. Paradoxically, motor axons show the most marked excitability changes (i.e. decreased superexcitability associated with slowing of inactivation of nodal voltage-gated sodium channel), despite that the sensory symptoms are the most important acute side-effect of oxaliplatin toxicity 33. Animal models have further indicated a crucial role of Nav1.6 62. A clinical report using blockade of peripheral myelinated A-fibers in combination with quantitative sensory testing (QST) implicated this this neuronal population as driving symptoms63.
Nerve imaging has also provided insights into PIPN. Nerve high-resolution ultrasound in oxaliplatin induced neuropathy showed normal cross sectional area in the non-entrapment sites and increased cross sectional area in the entrapment sites with no NCS evidence of entrapment 64, 65, which suggests these patients may be prone to develop entrapment neuropathies. Using magnetic resonance imaging diffusion tensor imaging, hypertrophy was observed in the DRGs, but not the proximal nerve segments, in a study of 20 patients with oxaliplatin-induced peripheral neuropathy 66.
While electrophysiology and imaging may provide valuable ancillary data regarding PIPN, it is worth noting that most patients with PIPN do not undergo this advanced testing. Most patients with PIPN are diagnosed clinically based on bedside signs and symptoms. Ancillary testing is not performed both due to the limited availability of the described modalities as well as the practicality and costs of performing many of these tests in a busy oncological practice. Depending on the practice, the described testing may be used in selected cases when the diagnosis is in doubt or if it is being performed for research purposes.
Risk Factors and Pharmacogenetics
The identification of risk factors that predict a worse neurological outcome in cisplatin-treated patients has been limited by the frequent use of the drug in combination with other neurotoxic agents (typically taxanes). In fact, while it is well known since the earliest stages of its clinical use that the severity of cisplatin neurotoxicity is dependent on both single and total dose administered 67, more refined and recent searches for additional risk factors failed to provide reliable evidence supporting the role of other predictors. Moreover, while subsequent studies confirmed that the incidence of cisplatin-induced peripheral neurotoxicity is related not only to dose per cycle and cumulative dose, but also to duration of infusion 68, these factors were not associated with long-term toxicity in a clinical follow-up study designed to evaluate adverse late effects in adult survivors of childhood cancer 69. Since cisplatin is used both in pediatric as well as in adult cancer treatment, age has been specifically investigated as a possible risk factor, without any evidence in support of this hypothesis 70.
Data are easier to interpret when oxaliplatin neurotoxicity is considered. Nearly all the patients treated with oxaliplatin experience acute, cold-related symptoms within hours from drug administration, although the severity of these symptoms might be different 57, 71, 72. However, the susceptibility of individual patients to chronic oxaliplatin-induced peripheral neurotoxicity is highly variable. This individual variability has been reproduced also in animal models, since different strains of mice chronically treated with oxaliplatin show a wide range of phenotypes in terms of severity as well as of type of neurological impairment 73.
This remarkable variability still needs to be fully explained because different studies have failed to unequivocally indicate which might be the risk factors or the predictors for the final neurological outcome in each patient. Apart from cumulative dose, which is clearly associated with more severe neurotoxicity 74-78, several other apparently obvious risk factors for peripheral nerve damage failed to demonstrate consistent results in different studies 78. This is the case for diabetes 79-81, age 76, 81-83, previous chemotherapy 74, while other risk factors less commonly associated to increased incidence of peripheral neuropathies, such as body mass index 80, 82 and smoking 80, 84, were occasionally reported.
The most recent systematic review on oxaliplatin neurotoxicity-associated risk factors 78 analyzed 322 papers focused on oxaliplatin-induced peripheral neurotoxicity and identified 72 studies on risk/prognostic factors. Among them, 15 reported clinical or patient-related factors (i.e patient demographics, baseline laboratory findings, comorbid disease, cancer diagnosis and nature and severity of neuropathy symptoms) and were further analyzed according to the STROBE Statement checklist 85. Among the selected studies, only 3 were prospective, and in most cases the distinction between acute and chronic symptoms induced by oxaliplatin was missing or unclear. Moreover, the methods used to assess the neurotoxic effect of oxaliplatin were highly variable across the 15 studies. Another major issue was related to the sample size of the studies, since only 4 of them investigated more than 200 patients, while 7 reported the results obtained in less than 60 patients. It is remarkable that the 2 largest studies (examining 1585 and 3359 patients, respectively) 86, 87, failed to observe any significant association with demographic or clinical risk factors.
The conclusions of the systematic review of the available data can be summarized in the following practical points: a) the identification of specific symptoms of acute neurotoxicity induced by oxaliplatin can be helpful to predict the onset of more severe chronic neurotoxicity 88, 89; b) there is no clear evidence for a different neurotoxicity between the 2 most commonly used chemotherapy regimens including oxaliplatin (i.e. FOLFOX and XELOX), despite different dose per cycle and interval between cycles 45; c) abnormal laboratory findings (i.e. low hemoglobin levels, hypoalbuminemia, hypomagnesemia, high serum chlorine levels) 81, 87 resulted to be associated with high risk of more severe neurotoxicity, but further research (particularly well designed prospective trials in homogeneous populations evaluated with proper neurological assessment) is required in order to prove their validity to predict chronic oxaliplatin-induced peripheral neurotoxicity 45.
Since demographic and clinical risk factors are still missing, a more sophisticated approach based on different pharmacogenetic methods have been attempted by several authors to solve the problem of early prediction of more severe neurological outcome in oxaliplatin-treated patients. Unfortunately, also using this approach no reliable predictors have been identified despite extensive investigation. This topic has been thoroughly reviewed considering several anticancer treatments 90-93. Platinum-based drugs (particularly oxaliplatin) are among the most frequently neurotoxic anticancer drugs investigated in pharmacogenetics studies, GSTP1 Ile105Val polymorphism being the most frequent candidate gene94, with very conflicting results in different analyses. Several other genetic associations have been reported, but also in these cases not subsequently validated. Although it is possible that no genetic predictors could eventually be found (similar analysis performed in mice where oxaliplatin administration resulted in different neurotoxicity failed to identify relevant genetic associations) 73, several major flaws have been identified in most of the reported clinical studies. To summarize, these are the major potential bias sources to be corrected in future studies: a) from the statistical standpoint, use of adequate sample size, adjustment for multiple comparison and comparison with appropriate validation cohorts; b) regarding patients assessment, identification of an accurate endpoint and selection of a reliable outcome measure; c) concerning biological samples and their analysis, selection of the most suitable approach (genome wide association studies, single nucleotide polymorphisms panels, gene expression studies) and validation of peripheral nervous system protein expression.
Prevention of Platinum-induced Peripheral Neurotoxicity: Clinical Trial Strategies
There are currently no evidence-based pharmacologic or supplement interventions for prevention of CIPN with platinum-based compounds (Table 3).95 Many agents have been tested to prevent CIPN, but have either shown no effect or there was an effect in a small sample study not confirmed in larger studies (for extensive review, see Hershman et al.95). As a recent example, calcium and magnesium infusions were not found to be beneficial in prevention of oxaliplatin CIPN in a large phase III randomized study96. The largest non-pharmacologic intervention study was Exercise for Cancer Patients (EXCAP), which was a randomized moderate-intensity, home-based, six-week progressive walking and resistance exercise program in patients undergoing taxane, platinum, or vinca alkaloid-based chemotherapy, as compared to usual care.97 Out of the 355 randomized patients, 39 (11%) patients received platinum-based chemotherapy. Patients in the exercise had significantly less temperature sensitivity and less sensory neuropathy symptoms. Since this was a secondary analysis of a previously conducted study on fatigue, a large prospective study is planned to confirm these findings. However, it does appear the exercise may have beneficial protective effects on CIPN from several chemotherapeutic agents including platinum-based drugs, although the numbers with this particular chemotherapeutic class were limited.
Table 3;
Intervention | Proposed mechanism of action |
Chemotherapy drug |
Sample size | Outcome measure |
Results | Reference |
---|---|---|---|---|---|---|
Prevention | ||||||
Amifostine (AM) | Cytoprotective agent via detoxification and ROS scavenging | Platinum | 242 AM:122 PL: 120 |
NCI-CTC after 6 cycles | NCI-CTC grade 1 toxicity: AM: 29%, PL: 31%, grade 2: AM: 29%, PL: 35%, grade 3: AM: 9%, PL: 15%, P=0.029 |
Kemp 1996145 |
Amifostine (AM) | Cytoprotective agent via detoxification and ROS scavenging | Carboplatin/paclitaxel | 187 AM: 93 PL: 94 |
NCI-CTC Grade 3-4 neurotoxicity | AM: 3.7% (95% CI, 2.1 to 5.3) PL: 7.2% (95% CI, 5.0 to 9.4) P=0.021 |
Lorusso 2003146 |
Amitiptyline (AMI) | Affects descending noradrenergic pathways | Vinca alcaloids, platinum, or taxanes | 99 AMI:54 PL: 45 |
Visual analogue scale (VAS) twice per week | No significant difference in neuropathy score at median of 19-21 wk follow up between AMI and PL groups P=n.s. |
Kautio 2009147 |
Calcium and magnesium (CaMg) infusions | Inhibits oxalate’s action on Na+ channels | Oxaliplatin | 353 CaMg/CaMg:118, CaMg/PL:116 PL/PL: 119 |
Sensory subscale of the EORTC QLQ-CIPN20 and CTCAE | CTCAE grade ≥ 2 neurotoxicity CaMg/CaMg: 43%, CaMg/PL: 46%, PL/PL: 45% P=n.s. |
Loprinzi 2013148 |
Diethyldithio-carbamate (DDTC) | Chelation tissue-bound platinum | Platinum | 214 DDTC: 106 PL: 108 |
NCI-CTC | DDTC: 13%, PL: 12%; P=n.s. |
Gandara 1995149 |
Glutathione (GSH) | Antioxidant; decreases DRG platinum | Paclitaxel/carboplatin | 185 GSH: 94 PL: 91 |
EORTC-QLQ-CIPN20 and CTCAE | Peripheral Neuropathy: P=0.21 CTCAE grade ≥ 2neurotoxicity: GSH: 38%, PL: 33% P=0.449 |
Leal 2013150 |
Glutathione (GSH) | Antioxidant; decreases DRG platinum | Cisplatin | 151 GSH: 74 PL: 77 |
NCI-CTC | GSH: 39%, PL: 49%; P = .22 | Smyth 1997151 |
Vitamin E (Vit. E) | Antioxidant | Taxane, cisplatin, carboplatin, oxaliplatin, or combination | 189 Vit. E: 96 PL: 93 |
CTCAE v 3.0 | Grade 2+: Vit E: 34%; PL: 29%; P=0.43 |
Kottschade 2011152 |
Recombinant human leukemia inhibitory factor (rhuLIF) | Neuroprotective growth factor effect | Carboplatin and paclitaxel | 117 rhuLIF 2 μg/kg: 36; rhuLIF 4 μg/kg: 39; PL: 42 |
CPNE score at cycle 4 | No significant differences seen in CPNE between time points for any treatment groups P=n.s. |
Davis 2005153 |
Calmangafodipir | Mimics the mitochondrial enzyme manganese superoxide dismutase by protecting cells from oxidative stress | FOLFOX-6 Folinic acid, 5-fluorouracil oxaliplatin | 173 PL= 60 Cal 2 μmol/kg=57; 5 μmol/kg=45, 10 μmol/kg=11 |
CTCAE; Leonard Scale Questionnaire; NCI-Sanofi ; cold allodynia test | Trend toward decreased physician graded neurotoxicity P=0.16 Decreased PRO symptoms P<0.01 |
Glimelius 2017154 |
Stop-and-Go Approach (earlier interruption of oxaliplatin and maintenance chemotherapy without it) | Less neurotoxic chemotherapy is administered before severe neurotoxicity develops | FOLFOX4 (Arm A) vs FOLFOX7 (Arm B) | 620 | NCI-Common Terminology Criteria for Adverse Events (CTCAE) | Grade 3 sensory neuropathy in 17.9% of the patients in arm A v 13.3% of patients in arm B P=0.12 |
Tournigand 2006155 |
Non Pharmacological Intervention Exercise (E) during chemotherapy vs Control group (C) |
Reduction of inflammation; affects sensory processing in the brain | Taxane-, platinum-, or Vinca alkaloid-based | 355 E: 155 C: 185 |
Patients reported grading of CIPN symptoms | Decreased temperature sensitivity P=0.045 Decreased sensory symptoms P=0.061 |
Kleckner, 201897 |
Treatment | ||||||
Gabapentin | Inhibition of voltage-dependent calcium channels in DRG | Vinca alkaloids, platinum, or taxanes | 115 G/P: 57 P/G: 58 |
NRS and ENS for ‘average’ daily pain at baseline and at the end of study | NSR scores for ‘average’ pain at study entry G/P; 4.3, P/G; 3.6 P=0.06 at 1st phase G/P; 3.3, P/G; 3.1 P=0.8 at 2nd phase G/P; 3.1 P/G 2.5 P=0.2 |
Rao, 2007156 |
Duloxetine (Dulox) | serotonin and norepinephrine reuptake inhibitor; sodium channel inhibition | Taxane or platinum | 231 Grp A: 115, dulox then cross over PL Grp B: 116, PL then cross over to dulox |
BPI-SF “average pain” item | Grp A average pain decreased more than Grp B P=0.003 |
Smith 2013157 |
Topical amitriptyline, ketamine, with or without Baclofen (BAK) | Amitriptyline: alters sodium channel and adenosine A receptor Ketamine: blocks NMDA receptors Baclofen: GABA agonist |
Vina alcaloids, platinum, taxanes, or thalidomide | 203 BAK: 101 PL: 102 |
Change in sensory subscale EORTC-QLQ-CIPN20 score at 4 wks | Mean change from baseline of sensory subscale for BAK (8.1) versus PL (3.8) P=0.053 |
Barton 2011158 |
Lamotrigine (Lam) | Inhibits sodium channels, blocking release of glutamate and aspartate | Vinca alcaloids, platinum, or taxanes | 131 Lam: 63; PL: 62 |
NRS and ENS at 10 wks | Mean NRS change from baseline for Lam (0.3) versus PL (0.5) P=0.56 Mean ENS change from baseline for Lam (0.4) versus PL (0.3) P=0.3 |
Rao 2008159 |
BPI-SF: Brief Pain Inventory-Short Form; CPNE: Standardized Composite Peripheral Nerve Electrophysiology Score; CTCAE: National Cancer Institute Common Terminology Criteria for Adverse Events; DRG: Dorsal Root Ganglia; ENS: Eastern Cooperative Oncology Group Neuropathy Scale; EORTC QLQ-CIPN20: European Organization for the Treatment of Cancer quality of life questionnaire-CIPN twenty item scale; NCI-CTC, National Cancer Institute Common Toxicity Criteria; NCI-Sanofi: oxaliplatin Sanofi questionnaire; NRS: Numerical Rating Scale; n.s: Not Statistically Significant; PL: Placebo; PRO: Patient Reported Outcome; ROS: Reactive Oxygen Species; VAS: Visual Analogue Scale;
Several ongoing prevention studies are evaluating the role of pharmacologic prevention strategies in PIPN. Oxidative stress from chemotherapy is the primary inducer of mitochondrial damage and neutralization of reactive oxygen species is a potential preventative strategy for platinum-induced peripheral neurotoxicity. Calmangafodipir, derived from a magnetic resonance imaging contrast agent known as mangafodipir, mimics the mitochondrial enzyme manganese superoxide dismutase, which reduces reactive oxygen species and subsequent nerve injury after platinum-based chemotherapy exposure. 98, 99 In a placebo-controlled, double-blinded randomized phase II study in patients with metastatic colorectal cancer, calmangafodipir reduced CIPN associated symptoms during and after treatment. 100 Two international trials, POLAR A and POLAR M, are currently evaluating the efficacy of calmangafodipir for the prevention of oxaliplatin-induced neuropathy in colorectal cancer patients. (Polar M: ). These phase III, multicenter, placebo-controlled trials are evaluating stage II and III patients in POLAR A, while POLAR M is being conducted in metastatic patients. Results are expected by the end of 2020.
While the published data on the analgesic effects of duloxetine for treatment of neuropathic pain is supported,101 emerging preclinical data suggest that duloxetine may also be effective for prevention of CIPN as well. A new NCI-funded clinical trial is currently undergoing protocol development to evaluate the efficacy of duloxetine for prevention of oxaliplatin-induced neuropathy (NCI, Ellen Smith PI).
Other interesting platinum-based peripheral neurotoxicity pathways being explored are in early stages of development. One pathway focuses on cellular transport of platinum-based compounds and aims to perform genetic or pharmacological knockout of transporters localized to the DRG in mice, such as OATP1B2 (OATP1B1 in humans), organic cation transporter novel type (OCTN2) protects against CIPN associated with and platinum-based drugs. 102, 103 A second molecule of interest is apyrimidinic endonuclease/redox effector factor-1 (APE1), which can be pharmacologically induced to increase DNA base excision repair, while simultaneously showing anti-cancer effects104. Both of these pathways are nearing translation into human clinical trials for CIPN prevention.
Treatment of Established Platinum-induced Peripheral Neurotoxicity: Clinical Trial Strategies
Prompt recognition of progressive CIPN symptoms remains the primary cornerstone of management of CIPN from these platinum-based agents, in order to reduce severity of symptom and risk of permanent injury. Early referral to physical therapy and rehab medicine is important for evaluation of functional deficits and falls risk. Depending on the severity of CIPN, dose reduction or dose holds are often necessary to manage the cumulative toxicity of all of these agents. The Stop and Go Approach which consists of stopping therapy before severe neurotoxicity develops, and later reintroducing oxlaliplatin did not yield significant differences in grade 3 or 4 CIPN. This approach is not routinely used in clinical care105. Additional management strategies include use of supportive care medications such as the traditional anti-neuropathic pain medications of antiepileptics and antidepressants. Despite the widespread use of these medications, to date duloxetine, a serotonin-norepinephrine reuptake inhibitor has been the only drug found to be associated with a significant reduction in neuropathic pain in CIPN in a randomized phase III intervention study (Table 3). 101 The mechanism of duloxetine-induced analgesia is due to the blocking of serotonin and norepinephrine transporters, as well as blocking of sodium channel currents and affecting the descending inhibitory pain neural networks 106-109 Additional novel therapeutic strategies are currently in development.110
Conclusions & Future Directions
Peripheral neurotoxicity from platinum-based compounds continues to be a serious dose-limiting side effect without any proven preventive therapies, despite intensive investigations over the past several decades. Given their efficacy to treat cancer, platinum-based chemotherapeutics are not going away anytime soon. Therefore, there will continue to be a critical need to better understand of PIPN, rigorously quantify its impairments, and develop effective preventative and treatment strategies. Furthermore, there continues to be active drug development for new platinum-based compounds, which is using several approaches111. First, platinum-compounds are being formed with novel attached side groups (for example, phenanthriplatin112), which cause diverse DNA adducts less susceptible to known mechanisms of drug resistance. Second, new platinum formulations have been developed that alter drug pharmacokinetics, such as liposomal cisplatin, which has been purported to be less neurotoxic113. Finally, platinum drugs are being conjugated with bioactive compounds in order to increase efficacy and better target cancer. The approach of combining platinum with a molecule that can be targeted to specific cancer has been used with microtubule-targeting drugs, such as brentuximab vedotin and ado-trastuzumab emtasnsine114, 115. Notably, in those cases, the attached compound was expected to reduce off-target effects; however, CIPN continues to be a serious side-effect, raising questions about specificity. As these newer formulations of platinum-based compounds enter the clinical practice, detailed characterization of their neurotoxicity will continue to be essential.
Acknowledgements
Grant Support: National Institutes of Health/National Cancer Institute: R01 CA 211887 (NPS)
References:
- 1.Rosenberg B, Van Camp L, Krigas T. Inhibition of Cell Division in Escherichia coli by Electrolysis Products from a Platinum Electrode. Nature. 1965;205:698–699. [DOI] [PubMed] [Google Scholar]
- 2.Rosenberg B, VanCamp L. The successful regression of large solid sarcoma 180 tumors by platinum compounds. Cancer Res. 1970;30:1799–1802. [PubMed] [Google Scholar]
- 3.Kelland L The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer. 2007;7:573–584. [DOI] [PubMed] [Google Scholar]
- 4.Gill JS, Windebank AJ. Cisplatin-induced apoptosis in rat dorsal root ganglion neurons is associated with attempted entry into the cell cycle. J Clin Invest. 1998;101:2842–2850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.McDonald ES, Windebank AJ. Cisplatin-induced apoptosis of DRG neurons involves bax redistribution and cytochrome c release but not fas receptor signaling. Neurobiol Dis. 2002;9:220–233. [DOI] [PubMed] [Google Scholar]
- 6.Podratz JL, Knight AM, Ta LE, et al. Cisplatin induced mitochondrial DNA damage in dorsal root ganglion neurons. Neurobiol Dis. 2011;41:661–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Park SB, Lin CS, Krishnan AV, Goldstein D, Friedlander ML, Kiernan MC. Oxaliplatin-induced neurotoxicity: changes in axonal excitability precede development of neuropathy. Brain. 2009;132:2712–2723. [DOI] [PubMed] [Google Scholar]
- 8.Pachman DR, Qin R, Seisler DK, et al. Clinical Course of Oxaliplatin-Induced Neuropathy: Results From the Randomized Phase III Trial N08CB (Alliance). Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015;33:3416–3422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ruggiero A, Trombatore G, Triarico S, et al. Platinum compounds in children with cancer: toxicity and clinical management. Anti-cancer drugs. 2013;24:1007–1019. [DOI] [PubMed] [Google Scholar]
- 10.Argyriou AA, Cavaletti G, Antonacopoulou A, et al. Voltage-gated sodium channel polymorphisms play a pivotal role in the development of oxaliplatin-induced peripheral neurotoxicity: results from a prospective multicenter study. Cancer. 2013;119:3570–3577. [DOI] [PubMed] [Google Scholar]
- 11.Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol. 2014;740:364–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Argyriou AA, Bruna J, Marmiroli P, Cavaletti G. Chemotherapy-induced peripheral neurotoxicity (CIPN): an update. Crit Rev Oncol Hematol. 2012;82:51–77. [DOI] [PubMed] [Google Scholar]
- 13.Argyriou AA, Zolota V, Kyriakopoulou O, Kalofonos HP. Toxic peripheral neuropathy associated with commonly used chemotherapeutic agents. Journal of B.U.ON. : official journal of the Balkan Union of Oncology. 2010;15:435–446. [PubMed] [Google Scholar]
- 14.Tofthagen C, McAllister RD, McMillan SC. Peripheral neuropathy in patients with colorectal cancer receiving oxaliplatin. Clinical journal of oncology nursing. 2011;15:182–188. [DOI] [PubMed] [Google Scholar]
- 15.Park SB, Lin CS, Krishnan AV, Goldstein D, Friedlander ML, Kiernan MC. Long-term neuropathy after oxaliplatin treatment: challenging the dictum of reversibility. The oncologist. 2011;16:708–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Arkenau HK. Capecitabine combined with oxaliplatin (CAPOX) in clinical practice: how significant is peripheral neuropathy? Annals of oncology : official journal of the European Society for Medical Oncology / ESMO. 2011;22:239–240; author reply 240–231. [DOI] [PubMed] [Google Scholar]
- 17.Amaraneni A, Seth A, Itawi EA, Chandana SR. Oxaliplatin-induced Lhermitte sign. A case report and review of literature. Clin Colorectal Cancer. 2014;13:257–259. [DOI] [PubMed] [Google Scholar]
- 18.Griffith KA, Zhu S, Johantgen M, et al. Oxaliplatin-Induced Peripheral Neuropathy and Identification of Unique Severity Groups in Colorectal Cancer. J Pain Symptom Manage. 2017;54:701–706 e701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yoon JY, Nam TS, Kim MK, et al. Acute inflammatory demyelinating polyradiculoneuropathy in a patient receiving oxaliplatin-based chemotherapy. Asia Pac J Clin Oncol. 2012;8:201–204. [DOI] [PubMed] [Google Scholar]
- 20.Verdonk RC, Enting RH, Janmaat M, Schroder CP, Sleijfer DT, Gietema JA. Weakness and numbness after chemotherapy for metastatic non-seminoma testis: a new neurological complication. Acta Oncol. 2008;47:1596–1598. [DOI] [PubMed] [Google Scholar]
- 21.Cicero G, Fulfaro F, Caraceni A, et al. A case of Guillain-Barre syndrome in a patient with non small cell lung cancer treated with chemotherapy. J Chemother. 2006;18:325–327. [DOI] [PubMed] [Google Scholar]
- 22.Tho LM, O’Leary CP, Horrocks I, Al-Ani A, Reed NS. Guillain-Barre syndrome occurring after adjuvant chemo-radiotherapy for endometrial cancer. Gynecol Oncol. 2006;100:615–617. [DOI] [PubMed] [Google Scholar]
- 23.Christodoulou C, Anastasopoulos D, Visvikis A, et al. Guillain-Barre syndrome in a patient with metastatic colon cancer receiving oxaliplatin-based chemotherapy. Anticancer Drugs. 2004;15:997–999. [DOI] [PubMed] [Google Scholar]
- 24.Johnson C, Pankratz VS, Velazquez AI, et al. Candidate pathway-based genetic association study of platinum and platinum-taxane related toxicity in a cohort of primary lung cancer patients. J Neurol Sci. 2015;349:124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Dermitzakis EV, Kimiskidis VK, Lazaridis G, et al. The impact of paclitaxel and carboplatin chemotherapy on the autonomous nervous system of patients with ovarian cancer. BMC Neurol. 2016;16:190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.van As JW, van den Berg H, van Dalen EC. Medical interventions for the prevention of platinum-induced hearing loss in children with cancer. Cochrane Database Syst Rev. 2014:CD009219. [DOI] [PubMed] [Google Scholar]
- 27.Knight KR, Kraemer DF, Neuwelt EA. Ototoxicity in children receiving platinum chemotherapy: underestimating a commonly occurring toxicity that may influence academic and social development. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2005;23:8588–8596. [DOI] [PubMed] [Google Scholar]
- 28.Knight KR, Kraemer DF, Winter C, Neuwelt EA. Early changes in auditory function as a result of platinum chemotherapy: use of extended high-frequency audiometry and evoked distortion product otoacoustic emissions. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2007;25:1190–1195. [DOI] [PubMed] [Google Scholar]
- 29.Blakley BW, Myers SF. Patterns of hearing loss resulting from cis-platinum therapy. Otolaryngology--head and neck surgery : official journal of American Academy of Otolaryngology-Head and Neck Surgery. 1993;109:385–391. [DOI] [PubMed] [Google Scholar]
- 30.Skinner R, Pearson AD, Amineddine HA, Mathias DB, Craft AW. Ototoxicity of cisplatinum in children and adolescents. British journal of cancer. 1990;61:927–931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Brock PR, Knight KR, Freyer DR, et al. Platinum-induced ototoxicity in children: a consensus review on mechanisms, predisposition, and protection, including a new International Society of Pediatric Oncology Boston ototoxicity scale. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30:2408–2417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.John T, Lomeli N, Bota DA. Systemic cisplatin exposure during infancy and adolescence causes impaired cognitive function in adulthood. Behavioural brain research. 2017;319:200–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Heide R, Bostock H, Ventzel L, et al. Axonal excitability changes and acute symptoms of oxaliplatin treatment: In vivo evidence for slowed sodium channel inactivation. Clin Neurophysiol. 2018;129:694–706. [DOI] [PubMed] [Google Scholar]
- 34.Lucchetta M, Lonardi S, Bergamo F, et al. Incidence of atypical acute nerve hyperexcitability symptoms in oxaliplatin-treated patients with colorectal cancer. Cancer Chemother Pharmacol. 2012;70:899–902. [DOI] [PubMed] [Google Scholar]
- 35.Tanishima H, Tominaga T, Kimura M, Maeda T, Shirai Y, Horiuchi T. Hyperacute peripheral neuropathy is a predictor of oxaliplatin-induced persistent peripheral neuropathy. Support Care Cancer. 2017;25:1383–1389. [DOI] [PubMed] [Google Scholar]
- 36.Reddy SM, Vergo MT, Paice JA, et al. Quantitative Sensory Testing at Baseline and During Cycle 1 Oxaliplatin Infusion Detects Subclinical Peripheral Neuropathy and Predicts Clinically Overt Chronic Neuropathy in Gastrointestinal Malignancies. Clin Colorectal Cancer. 2016;15:37–46. [DOI] [PubMed] [Google Scholar]
- 37.Gebremedhn EG, Shortland PJ, Mahns DA. The incidence of acute oxaliplatin-induced neuropathy and its impact on treatment in the first cycle: a systematic review. BMC cancer. 2018;18:410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Glendenning JL, Barbachano Y, Norman AR, Dearnaley DP, Horwich A, Huddart RA. Long-term neurologic and peripheral vascular toxicity after chemotherapy treatment of testicular cancer. Cancer. 2010;116:2322–2331. [DOI] [PubMed] [Google Scholar]
- 39.Albers JW, Chaudhry V, Cavaletti G, Donehower RC. Interventions for preventing neuropathy caused by cisplatin and related compounds. Cochrane Database Syst Rev. 2014:CD005228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Krishnan AV, Goldstein D, Friedlander M, Kiernan MC. Oxaliplatin-induced neurotoxicity and the development of neuropathy. Muscle Nerve. 2005;32:51–60. [DOI] [PubMed] [Google Scholar]
- 41.Pietrangeli A, Leandri M, Terzoli E, Jandolo B, Garufi C. Persistence of high-dose oxaliplatin-induced neuropathy at long-term follow-up. Eur Neurol. 2006;56:13–16. [DOI] [PubMed] [Google Scholar]
- 42.Matsumoto S, Nishimura T, Kanai M, et al. Safety and efficacy of modified FOLFOX6 for treatment of metastatic or locally advanced colorectal cancer. A single-institution outcome study. Chemotherapy. 2008;54:395–403. [DOI] [PubMed] [Google Scholar]
- 43.Storey DJ, Sakala M, McLean CM, et al. Capecitabine combined with oxaliplatin (CapOx) in clinical practice: how significant is peripheral neuropathy? Ann Oncol. 2010;21:1657–1661. [DOI] [PubMed] [Google Scholar]
- 44.Gurney H, Crowther D, Anderson H, et al. Five year follow-up and dose delivery analysis of cisplatin, iproplatin or carboplatin in combination with cyclophosphamide in advanced ovarian carcinoma. Ann Oncol. 1990;1:427–433. [PubMed] [Google Scholar]
- 45.Beijers AJ, Mols F, Vreugdenhil G. A systematic review on chronic oxaliplatin-induced peripheral neuropathy and the relation with oxaliplatin administration. Support Care Cancer. 2014;22:1999–2007. [DOI] [PubMed] [Google Scholar]
- 46.Adjuvant chemotherapy with oxaliplatin, in combination with fluorouracil plus leucovorin prolongs disease-free survival, but causes more adverse events in people with stage II or III colon cancer Abstracted from: <Andre T, Boni C, Mounedji-Boudiaf L, et al. Multicenter international study of oxaliplatin/5-fluorouracil/leucovorin in the adjuvant treatment of colon cancer (MOSAIC) investigators. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N Engl J Med 2004;350:2343–51. Cancer treatment reviews. 2004;30:711–713. [DOI] [PubMed] [Google Scholar]
- 47.Briani C, Argyriou AA, Izquierdo C, et al. Long-term course of oxaliplatin-induced polyneuropathy: a prospective 2-year follow-up study. J Peripher Nerv Syst. 2014;19:299–306. [DOI] [PubMed] [Google Scholar]
- 48.Kokotis P, Schmelz M, Kostouros E, Karandreas N, Dimopoulos MA. Oxaliplatin-Induced Neuropathy: A Long-Term Clinical and Neurophysiologic Follow-Up Study. Clin Colorectal Cancer. 2016;15:e133–140. [DOI] [PubMed] [Google Scholar]
- 49.Park SB, Goldstein D, Krishnan AV, et al. Chemotherapy-induced peripheral neurotoxicity: a critical analysis. CA: a cancer journal for clinicians. 2013;63:419–437. [DOI] [PubMed] [Google Scholar]
- 50.Mollman JE, Glover DJ, Hogan WM, Furman RE. Cisplatin neuropathy. Risk factors, prognosis, and protection by WR-2721. Cancer. 1988;61:2192–2195. [DOI] [PubMed] [Google Scholar]
- 51.Burakgazi AZ, Messersmith W, Vaidya D, Hauer P, Hoke A, Polydefkis M. Longitudinal assessment of oxaliplatin-induced neuropathy. Neurology. 2011;77:980–986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kroigard T, Schroder HD, Qvortrup C, et al. Characterization and diagnostic evaluation of chronic polyneuropathies induced by oxaliplatin and docetaxel comparing skin biopsy to quantitative sensory testing and nerve conduction studies. European journal of neurology : the official journal of the European Federation of Neurological Societies. 2014;21:623–629. [DOI] [PubMed] [Google Scholar]
- 53.Kandula T, Farrar MA, Cohn RJ, et al. Chemotherapy-Induced Peripheral Neuropathy in Long-term Survivors of Childhood Cancer: Clinical, Neurophysiological, Functional, and Patient-Reported Outcomes. JAMA Neurol. 2018;75:980–988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Krarup-Hansen A, Helweg-Larsen S, Schmalbruch H, Rorth M, Krarup C. Neuronal involvement in cisplatin neuropathy: prospective clinical and neurophysiological studies. Brain : a journal of neurology. 2007;130:1076–1088. [DOI] [PubMed] [Google Scholar]
- 55.Argyriou AA, Polychronopoulos P, Iconomou G, et al. Incidence and characteristics of peripheral neuropathy during oxaliplatin-based chemotherapy for metastatic colon cancer. Acta oncologica (Stockholm, Sweden). 2007;46:1131–1137. [DOI] [PubMed] [Google Scholar]
- 56.Mileshkin L, Stark R, Day B, Seymour JF, Zeldis JB, Prince HM. Development of neuropathy in patients with myeloma treated with thalidomide: patterns of occurrence and the role of electrophysiologic monitoring. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2006;24:4507–4514. [DOI] [PubMed] [Google Scholar]
- 57.Velasco R, Bruna J, Briani C, et al. Early predictors of oxaliplatin-induced cumulative neuropathy in colorectal cancer patients. J Neurol Neurosurg Psychiatry. 2014;85:392–398. [DOI] [PubMed] [Google Scholar]
- 58.Dalla Torre C, Zambello R, Cacciavillani M, et al. Lenalidomide long-term neurotoxicity: Clinical and neurophysiologic prospective study. Neurology. 2016;87:1161–1166. [DOI] [PubMed] [Google Scholar]
- 59.Kiernan MC, Burke D, Andersen KV, Bostock H. Multiple measures of axonal excitability: a new approach in clinical testing. Muscle Nerve. 2000;23:399–409. [DOI] [PubMed] [Google Scholar]
- 60.Wilson RH, Lehky T, Thomas RR, Quinn MG, Floeter MK, Grem JL. Acute oxaliplatin-induced peripheral nerve hyperexcitability. J Clin Oncol. 2002;20:1767–1774. [DOI] [PubMed] [Google Scholar]
- 61.Krishnan AV, Goldstein D, Friedlander M, Kiernan MC. Oxaliplatin and axonal Na+ channel function in vivo. Clin Cancer Res. 2006;12:4481–4484. [DOI] [PubMed] [Google Scholar]
- 62.Deuis JR, Zimmermann K, Romanovsky AA, et al. An animal model of oxaliplatin-induced cold allodynia reveals a crucial role for Nav1.6 in peripheral pain pathways. Pain. 2013;154:1749–1757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Forstenpointner J, Oberlojer VC, Naleschinski D, et al. A-Fibers Mediate Cold Hyperalgesia in Patients with Oxaliplatin-Induced Neuropathy. Pain Pract. 2018;18:758–767. [DOI] [PubMed] [Google Scholar]
- 64.Briani C, Campagnolo M, Lucchetta M, et al. Ultrasound assessment of oxaliplatin-induced neuropathy and correlations with neurophysiologic findings. European journal of neurology : the official journal of the European Federation of Neurological Societies. 2013;20:188–192. [DOI] [PubMed] [Google Scholar]
- 65.Pitarokoili K, Hoffken N, Lonneker N, et al. Prospective Study of the Clinical, Electrophysiologic, and Sonographic Characteristics of Oxaliplatin-Induced Neuropathy. Journal of neuroimaging : official journal of the American Society of Neuroimaging. 2019;29:133–139. [DOI] [PubMed] [Google Scholar]
- 66.Apostolidis L, Schwarz D, Xia A, et al. Dorsal root ganglia hypertrophy as in vivo correlate of oxaliplatin-induced polyneuropathy. PloS one. 2017;12:e0183845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Cavaletti G, Marzorati L, Bogliun G, et al. Cisplatin-induced peripheral neurotoxicity is dependent on total-dose intensity and single-dose intensity. Cancer. 1992;69:203–207. [DOI] [PubMed] [Google Scholar]
- 68.Ewertz M, Qvortrup C, Eckhoff L. Chemotherapy-induced peripheral neuropathy in patients treated with taxanes and platinum derivatives. Acta Oncol. 2015;54:587–591. [DOI] [PubMed] [Google Scholar]
- 69.Ness KK, Jones KE, Smith WA, et al. Chemotherapy-related neuropathic symptoms and functional impairment in adult survivors of extracranial solid tumors of childhood: results from the St. Jude Lifetime Cohort Study. Arch Phys Med Rehabil. 2013;94:1451–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Argyriou AA, Polychronopoulos P, Koutras A, et al. Is advanced age associated with increased incidence and severity of chemotherapy-induced peripheral neuropathy? Support Care Cancer. 2006;14:223–229. [DOI] [PubMed] [Google Scholar]
- 71.Argyriou AA. Updates on Oxaliplatin-Induced Peripheral Neurotoxicity (OXAIPN). Toxics. 2015;3:187–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Argyriou AA, Cavaletti G, Briani C, et al. Clinical pattern and associations of oxaliplatin acute neurotoxicity: a prospective study in 170 patients with colorectal cancer. Cancer. 2013;119:438–444. [DOI] [PubMed] [Google Scholar]
- 73.Marmiroli P, Riva B, Pozzi E, et al. Susceptibility of different mouse strains to oxaliplatin peripheral neurotoxicity: Phenotypic and genotypic insights. PLoS One. 2017;12:e0186250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Besora S, Santos C, Izquierdo C, Martinez-Villacampa MM, Bruna J, Velasco R. Rechallenge with oxaliplatin and peripheral neuropathy in colorectal cancer patients. J Cancer Res Clin Oncol. 2018;144:1793–1801. [DOI] [PubMed] [Google Scholar]
- 75.Hsu SY, Huang WS, Lee SH, et al. Incidence, severity, longitudinal trends and predictors of acute and chronic oxaliplatin-induced peripheral neuropathy in Taiwanese patients with colorectal cancer. Eur J Cancer Care (Engl). 2019;28:e12976. [DOI] [PubMed] [Google Scholar]
- 76.Yamaguchi K, Kusaba H, Makiyama A, et al. The risk factors for oxaliplatin-induced peripheral sensory neuropathy and thrombocytopenia in advanced gastric cancer. Cancer Chemother Pharmacol. 2018;82:625–633. [DOI] [PubMed] [Google Scholar]
- 77.Kerckhove N, Collin A, Condé S, Chaleteix C, Pezet D, Balayssac D. Long-Term Effects, Pathophysiological Mechanisms, and Risk Factors of Chemotherapy-Induced Peripheral Neuropathies: A Comprehensive Literature Review. Front Pharmacol. 2017;8:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Pulvers JN, Marx G. Factors associated with the development and severity of oxaliplatin-induced peripheral neuropathy: a systematic review. Asia Pac J Clin Oncol. 2017. [DOI] [PubMed] [Google Scholar]
- 79.Bano N, Ikram R. Effect of diabetes on neurological adverse effects and chemotherapy induced peripheral neuropathy in advanced colorectal cancer patients treated with different FOLFOX regimens. Pak J Pharm Sci. 2019;32:125–130. [PubMed] [Google Scholar]
- 80.Ottaiano A, Nappi A, Tafuto S, et al. Diabetes and Body Mass Index Are Associated with Neuropathy and Prognosis in Colon Cancer Patients Treated with Capecitabine and Oxaliplatin Adjuvant Chemotherapy. Oncology. 2016;90:36–42. [DOI] [PubMed] [Google Scholar]
- 81.Vincenzi B, Frezza AM, Schiavon G, et al. Identification of clinical predictive factors of oxaliplatin-induced chronic peripheral neuropathy in colorectal cancer patients treated with adjuvant Folfox IV. Support Care Cancer. 2013;21:1313–1319. [DOI] [PubMed] [Google Scholar]
- 82.Alejandro LM, Behrendt CE, Chen K, Openshaw H, Shibata S. Predicting acute and persistent neuropathy associated with oxaliplatin. Am J Clin Oncol. 2013;36:331–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Argyriou AA, Briani C, Cavaletti G, et al. Advanced age and liability to oxaliplatin-induced peripheral neuropathy: post hoc analysis of a prospective study. Eur J Neurol. 2013;20:788–794. [DOI] [PubMed] [Google Scholar]
- 84.Farker K, Merkel U, Wedding U, Hippius M, Höffken K, Hoffmann A. Chronomodulated chemotherapy with oxaliplatin, 5-FU and sodium folinate in metastatic gastrointestinal cancer patients: original analysis of non-hematological toxicity and patient characteristics in a pilot investigation. Int J Clin Pharmacol Ther. 2006;44:31–37. [DOI] [PubMed] [Google Scholar]
- 85.von Elm E, Altman DG, Egger M, et al. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) Statement: guidelines for reporting observational studies. Int J Surg. 2014;12:1495–1499. [DOI] [PubMed] [Google Scholar]
- 86.Ramanathan RK, Rothenberg ML, de Gramont A, et al. Incidence and evolution of oxaliplatin-induced peripheral sensory neuropathy in diabetic patients with colorectal cancer: a pooled analysis of three phase III studies. Ann Oncol. 2010;21:754–758. [DOI] [PubMed] [Google Scholar]
- 87.Sugihara K, Ohtsu A, Shimada Y, et al. Analysis of neurosensory adverse events induced by FOLFOX4 treatment in colorectal cancer patients: a comparison between two Asian studies and four Western studies. Cancer Med. 2012;1:198–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Mols F, Beijers T, Lemmens V, van den Hurk CJ, Vreugdenhil G, van de Poll-Franse LV. Chemotherapy-induced neuropathy and its association with quality of life among 2- to 11-year colorectal cancer survivors: results from the population-based PROFILES registry. J Clin Oncol. 2013;31:2699–2707. [DOI] [PubMed] [Google Scholar]
- 89.Tofthagen C, Donovan KA, Morgan MA, Shibata D, Yeh Y. Oxaliplatin-induced peripheral neuropathy’s effects on health-related quality of life of colorectal cancer survivors. Support Care Cancer. 2013;21:3307–3313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Terrazzino S, Argyriou AA, Cargnin S, et al. Genetic determinants of chronic oxaliplatin-induced peripheral neurotoxicity: a genome-wide study replication and meta-analysis. J Peripher Nerv Syst. 2015. [DOI] [PubMed] [Google Scholar]
- 91.Cavaletti G, Alberti P, Marmiroli P. Chemotherapy-induced peripheral neurotoxicity in the era of pharmacogenomics. Lancet Oncol. 2011;12:1151–1161. [DOI] [PubMed] [Google Scholar]
- 92.Argyriou AA, Bruna J, Genazzani AA, Cavaletti G. Chemotherapy-induced peripheral neurotoxicity: management informed by pharmacogenetics. Nat Rev Neurol. 2017;13:492–504. [DOI] [PubMed] [Google Scholar]
- 93.Mahmoudpour SH, Bandapalli OR, da Silva Filho MI, et al. Chemotherapy-induced peripheral neuropathy: evidence from genome-wide association studies and replication within multiple myeloma patients. BMC Cancer. 2018;18:820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lecomte T, Landi B, Beaune P, Laurent-Puig P, Loriot MA. Glutathione S-transferase P1 polymorphism (Ile105Val) predicts cumulative neuropathy in patients receiving oxaliplatin-based chemotherapy. Clinical cancer research : an official journal of the American Association for Cancer Research. 2006;12:3050–3056. [DOI] [PubMed] [Google Scholar]
- 95.Hershman DL, Lacchetti C, Dworkin RH, et al. Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2014;32:1941–1967. [DOI] [PubMed] [Google Scholar]
- 96.Loprinzi CL, Qin R, Dakhil SR, et al. Phase III randomized, placebo-controlled, double-blind study of intravenous calcium and magnesium to prevent oxaliplatin-induced sensory neurotoxicity (N08CB/Alliance). J Clin Oncol. 2014;32:997–1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kleckner IR, Kamen C, Gewandter JS, et al. Effects of exercise during chemotherapy on chemotherapy-induced peripheral neuropathy: a multicenter, randomized controlled trial. Support Care Cancer. 2018;26:1019–1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Karlsson JO, Ignarro LJ, Lundstrom I, Jynge P, Almen T. Calmangafodipir [Ca4Mn(DPDP)5], mangafodipir (MnDPDP) and MnPLED with special reference to their SOD mimetic and therapeutic properties. Drug Discov Today. 2015;20:411–421. [DOI] [PubMed] [Google Scholar]
- 99.Karlsson JO, Kurz T, Flechsig S, Nasstrom J, Andersson RG. Superior therapeutic index of calmangafodipir in comparison to mangafodipir as a chemotherapy adjunct. Transl Oncol. 2012;5:492–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Glimelius B, Manojlovic N, Pfeiffer P, et al. Persistent prevention of oxaliplatin-induced peripheral neuropathy using calmangafodipir (PledOx((R))): a placebo-controlled randomised phase II study (PLIANT). Acta Oncol. 2018;57:393–402. [DOI] [PubMed] [Google Scholar]
- 101.Smith EM, Pang H, Cirrincione C, et al. Effect of duloxetine on pain, function, and quality of life among patients with chemotherapy-induced painful peripheral neuropathy: a randomized clinical trial. Jama. 2013;309:1359–1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Fujita S, Hirota T, Sakiyama R, Baba M, Ieiri I. Identification of drug transporters contributing to oxaliplatin-induced peripheral neuropathy. J Neurochem. 2019;148:373–385. [DOI] [PubMed] [Google Scholar]
- 103.Sprowl JA, Ong SS, Gibson AA, et al. A phosphotyrosine switch regulates organic cation transporters. Nat Commun. 2016;7:10880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kelley MR, Jiang Y, Guo C, Reed A, Meng H, Vasko MR. Role of the DNA base excision repair protein, APE1 in cisplatin, oxaliplatin, or carboplatin induced sensory neuropathy. PLoS One. 2014;9:e106485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Tournigand C, Cervantes A, Figer A, et al. OPTIMOX1: a randomized study of FOLFOX4 or FOLFOX7 with oxaliplatin in a stop-and-Go fashion in advanced colorectal cancer--a GERCOR study. J Clin Oncol. 2006;24:394–400. [DOI] [PubMed] [Google Scholar]
- 106.Bymaster FP, Dreshfield-Ahmad LJ, Threlkeld PG, et al. Comparative affinity of duloxetine and venlafaxine for serotonin and norepinephrine transporters in vitro and in vivo, human serotonin receptor subtypes, and other neuronal receptors. Neuropsychopharmacol. 2001;25:871–880. [DOI] [PubMed] [Google Scholar]
- 107.Wang SY, Calderon J, Kuo Wang G. Block of neuronal Na+ channels by antidepressant duloxetine in a state-dependent manner. Anesthesiology. 2010;113:655–665. [DOI] [PubMed] [Google Scholar]
- 108.Wang CF, Russell G, Strichartz GR, Wang GK. The Local and Systemic Actions of Duloxetine in Allodynia and Hyperalgesia Using a Rat Skin Incision Pain Model. Anesthesia and analgesia. 2015;121:532–544. [DOI] [PubMed] [Google Scholar]
- 109.Skljarevski V, Zhang S, Iyengar S, et al. Efficacy of Duloxetine in Patients with Chronic Pain Conditions. Curr Drug ther. 2011;6:296–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Dorsey SG, Kleckner IR, Barton D, et al. NCI Clinical Trials Planning Meeting for prevention and treatment of chemotherapy-induced peripheral neuropathy. Journal of the National Cancer Institute. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Ghosh S Cisplatin: The first metal based anticancer drug. Bioorg Chem. 2019;88:102925. [DOI] [PubMed] [Google Scholar]
- 112.Gregory MT, Park GY, Johnstone TC, Lee YS, Yang W, Lippard SJ. Structural and mechanistic studies of polymerase eta bypass of phenanthriplatin DNA damage. Proc Natl Acad Sci U S A. 2014;111:9133–9138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Xu B, Zeng M, Zeng J, Feng J, Yu L. Meta-analysis of clinical trials comparing the efficacy and safety of liposomal cisplatin versus conventional nonliposomal cisplatin in nonsmall cell lung cancer (NSCLC) and squamous cell carcinoma of the head and neck (SCCHN). Medicine (Baltimore). 2018;97:e13169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Gopal AK, Ramchandren R, O’Connor OA, et al. Safety and efficacy of brentuximab vedotin for Hodgkin lymphoma recurring after allogeneic stem cell transplantation. Blood. 2012;120:560–568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Krop IE, Modi S, LoRusso PM, et al. Phase 1b/2a study of trastuzumab emtansine (T-DM1), paclitaxel, and pertuzumab in HER2-positive metastatic breast cancer. Breast Cancer Res. 2016;18:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Swenerton K, Jeffrey J, Stuart G, et al. Cisplatin-cyclophosphamide versus carboplatin-cyclophosphamide in advanced ovarian cancer: a randomized phase III study of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 1992;10:718–726. [DOI] [PubMed] [Google Scholar]
- 117.Kemp G, Rose P, Lurain J, et al. Amifostine pretreatment for protection against cyclophosphamide-induced and cisplatin-induced toxicities: results of a randomized control trial in patients with advanced ovarian cancer. J Clin Oncol. 1996;14:2101–2112. [DOI] [PubMed] [Google Scholar]
- 118.Wozniak AJ, Crowley JJ, Balcerzak SP, et al. Randomized trial comparing cisplatin with cisplatin plus vinorelbine in the treatment of advanced non-small-cell lung cancer: a Southwest Oncology Group study. J Clin Oncol. 1998;16:2459–2465. [DOI] [PubMed] [Google Scholar]
- 119.Sutton G, Brunetto VL, Kilgore L, et al. A phase III trial of ifosfamide with or without cisplatin in carcinosarcoma of the uterus: A Gynecologic Oncology Group Study. Gynecol Oncol. 2000;79:147–153. [DOI] [PubMed] [Google Scholar]
- 120.Gatzemeier U, von Pawel J, Gottfried M, et al. Phase III comparative study of high-dose cisplatin versus a combination of paclitaxel and cisplatin in patients with advanced non-small-cell lung cancer. J Clin Oncol. 2000;18:3390–3399. [DOI] [PubMed] [Google Scholar]
- 121.Wachters FM, Van Putten JW, Kramer H, et al. First-line gemcitabine with cisplatin or epirubicin in advanced non-small-cell lung cancer: a phase III trial. Br J Cancer. 2003;89:1192–1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Fleming GF, Brunetto VL, Cella D, et al. Phase III trial of doxorubicin plus cisplatin with or without paclitaxel plus filgrastim in advanced endometrial carcinoma: a Gynecologic Oncology Group Study. J Clin Oncol. 2004;22:2159–2166. [DOI] [PubMed] [Google Scholar]
- 123.Al-Batran SE, Hartmann JT, Probst S, et al. Phase III trial in metastatic gastroesophageal adenocarcinoma with fluorouracil, leucovorin plus either oxaliplatin or cisplatin: a study of the Arbeitsgemeinschaft Internistische Onkologie. J Clin Oncol. 2008;26:1435–1442. [DOI] [PubMed] [Google Scholar]
- 124.Homesley HD, Filiaci V, Gibbons SK, et al. A randomized phase III trial in advanced endometrial carcinoma of surgery and volume directed radiation followed by cisplatin and doxorubicin with or without paclitaxel: A Gynecologic Oncology Group study. Gynecol Oncol. 2009;112:543–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Chen Y, Sun Y, Liang SB, et al. Progress report of a randomized trial comparing long-term survival and late toxicity of concurrent chemoradiotherapy with adjuvant chemotherapy versus radiotherapy alone in patients with stage III to IVB nasopharyngeal carcinoma from endemic regions of China. Cancer. 2013;119:2230–2238. [DOI] [PubMed] [Google Scholar]
- 126.Friedrich C, von Bueren AO, von Hoff K, et al. Treatment of adult nonmetastatic medulloblastoma patients according to the paediatric HIT 2000 protocol: a prospective observational multicentre study. Eur J Cancer. 2013;49:893–903. [DOI] [PubMed] [Google Scholar]
- 127.Conroy T, Galais MP, Raoul JL, et al. Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): final results of a randomised, phase 2/3 trial. Lancet Oncol. 2014;15:305–314. [DOI] [PubMed] [Google Scholar]
- 128.Yamada Y, Higuchi K, Nishikawa K, et al. Phase III study comparing oxaliplatin plus S-1 with cisplatin plus S-1 in chemotherapy-naive patients with advanced gastric cancer. Ann Oncol. 2015;26:141–148. [DOI] [PubMed] [Google Scholar]
- 129.Bando H, Yamada Y, Tanabe S, et al. Efficacy and safety of S-1 and oxaliplatin combination therapy in elderly patients with advanced gastric cancer. Gastric Cancer. 2016;19:919–926. [DOI] [PubMed] [Google Scholar]
- 130.Fung C, Sesso HD, Williams AM, et al. Multi-Institutional Assessment of Adverse Health Outcomes Among North American Testicular Cancer Survivors After Modern Cisplatin-Based Chemotherapy. J Clin Oncol. 2017;35:1211–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Ramaswamy A, Ostwal V, Pinninti R, et al. Gemcitabine-cisplatin versus gemcitabine-oxaliplatin doublet chemotherapy in advanced gallbladder cancers: a match pair analysis. J Hepatobiliary Pancreat Sci. 2017;24:262–267. [DOI] [PubMed] [Google Scholar]
- 132.Giacchetti S, Perpoint B, Zidani R, et al. Phase III multicenter randomized trial of oxaliplatin added to chronomodulated fluorouracil-leucovorin as first-line treatment of metastatic colorectal cancer. J Clin Oncol. 2000;18:136–147. [DOI] [PubMed] [Google Scholar]
- 133.de Gramont A, Figer A, Seymour M, et al. Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol. 2000;18:2938–2947. [DOI] [PubMed] [Google Scholar]
- 134.Rothenberg ML, Oza AM, Bigelow RH, et al. Superiority of oxaliplatin and fluorouracil-leucovorin compared with either therapy alone in patients with progressive colorectal cancer after irinotecan and fluorouracil-leucovorin: interim results of a phase III trial. J Clin Oncol. 2003;21:2059–2069. [DOI] [PubMed] [Google Scholar]
- 135.Andre T, Boni C, Mounedji-Boudiaf L, et al. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N Engl J Med. 2004;350:2343–2351. [DOI] [PubMed] [Google Scholar]
- 136.Land SR, Kopec JA, Cecchini RS, et al. Neurotoxicity from oxaliplatin combined with weekly bolus fluorouracil and leucovorin as surgical adjuvant chemotherapy for stage II and III colon cancer: NSABP C-07. J Clin Oncol. 2007;25:2205–2211. [DOI] [PubMed] [Google Scholar]
- 137.Cassidy J, Clarke S, Diaz-Rubio E, et al. Randomized phase III study of capecitabine plus oxaliplatin compared with fluorouracil/folinic acid plus oxaliplatin as first-line therapy for metastatic colorectal cancer. J Clin Oncol. 2008;26:2006–2012. [DOI] [PubMed] [Google Scholar]
- 138.Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36–46. [DOI] [PubMed] [Google Scholar]
- 139.Allegra CJ, Yothers G, O’Connell MJ, et al. Initial safety report of NSABP C-08: A randomized phase III study of modified FOLFOX6 with or without bevacizumab for the adjuvant treatment of patients with stage II or III colon cancer. J Clin Oncol. 2009;27:3385–3390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Poplin E, Feng Y, Berlin J, et al. Phase III, randomized study of gemcitabine and oxaliplatin versus gemcitabine (fixed-dose rate infusion) compared with gemcitabine (30-minute infusion) in patients with pancreatic carcinoma E6201: a trial of the Eastern Cooperative Oncology Group. J Clin Oncol. 2009;27:3778–3785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Qvortrup C, Jensen BV, Fokstuen T, et al. A randomized study comparing short-time infusion of oxaliplatin in combination with capecitabine XELOX(30) and chronomodulated XELOX(30) as first-line therapy in patients with advanced colorectal cancer. Ann Oncol. 2010;21:87–91. [DOI] [PubMed] [Google Scholar]
- 142.Argyriou AA, Velasco R, Briani C, et al. Peripheral neurotoxicity of oxaliplatin in combination with 5-fluorouracil (FOLFOX) or capecitabine (XELOX): a prospective evaluation of 150 colorectal cancer patients. Ann Oncol. 2012;23:3116–3122. [DOI] [PubMed] [Google Scholar]
- 143.Lonardi S, Sobrero A, Rosati G, et al. Phase III trial comparing 3–6 months of adjuvant FOLFOX4/XELOX in stage II-III colon cancer: safety and compliance in the TOSCA trial. Ann Oncol. 2016;27:2074–2081. [DOI] [PubMed] [Google Scholar]
- 144.Iveson TJ, Kerr RS, Saunders MP, et al. 3 versus 6 months of adjuvant oxaliplatin-fluoropyrimidine combination therapy for colorectal cancer (SCOT): an international, randomised, phase 3, non-inferiority trial. Lancet Oncol. 2018;19:562–578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Kemp G, Rose P, Lurain J, et al. Amifostine pretreatment for protection against cyclophosphamide-induced and cisplatin-induced toxicities: results of a randomized control trial in patients with advanced ovarian cancer. Journal of Clinical Oncology. 1996;14:2101–2112. [DOI] [PubMed] [Google Scholar]
- 146.Lorusso D, Ferrandina G, Greggi S, et al. Phase III multicenter randomized trial of amifostine as cytoprotectant in first-line chemotherapy in ovarian cancer patients. Annals of Oncology. 2003;14:1086–1093. [DOI] [PubMed] [Google Scholar]
- 147.Kautio A-L, HAANPÄÄ M, Leminen A, Kalso E, Kautiainen H, Saarto T. Amitriptyline in the prevention of chemotherapy-induced neuropathic symptoms. Anticancer Research. 2009;29:2601–2606. [PubMed] [Google Scholar]
- 148.Loprinzi CL, Qin R, Dakhil SR, et al. Phase III randomized, placebo (PL)-controlled, double-blind study of intravenous calcium/magnesium (CaMg) to prevent oxaliplatin-induced sensory neurotoxicity (sNT), N08CB: An alliance for clinical trials in oncology study. Journal of Clinical Oncology. 2013;31:3501–3501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Gandara DR, Nahhas WA, Adelson MD, et al. Randomized placebo-controlled multicenter evaluation of diethyldithiocarbamate for chemoprotection against cisplatin-induced toxicities. Journal of Clinical Oncology. 1995;13:490–496. [DOI] [PubMed] [Google Scholar]
- 150.Leal AD, Qin R, Atherton PJ, et al. NCCTG N08CA (Alliance): The use of Glutathione for Prevention of Paclitaxel/Carboplatin Induced Peripheral Neuropathy: A Phase III Randomized, Double-Blind Placebo-Controlled Study. Cancer. 2014;120:1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Smyth J, Bowman A, Perren T, et al. Glutathione reduces the toxicity and improves quality of life of women diagnosed with ovarian cancer treated with cisplatin: results of a double-blind, randomised trial. Annals of oncology. 1997;8:569–573. [DOI] [PubMed] [Google Scholar]
- 152.Kottschade LA, Sloan JA, Mazurczak MA, et al. The use of vitamin E for the prevention of chemotherapy-induced peripheral neuropathy: results of a randomized phase III clinical trial. Supportive Care in Cancer. 2011;19:1769–1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Davis ID, Kiers L, MacGregor L, et al. A randomized, double-blinded, placebo-controlled phase II trial of recombinant human leukemia inhibitory factor (rhuLIF, emfilermin, AM424) to prevent chemotherapy-induced peripheral neuropathy. Clinical Cancer Research. 2005;11:1890–1898. [DOI] [PubMed] [Google Scholar]
- 154.Glimelius B, Manojlovic N, Pfeiffer P, et al. Persistent prevention of oxaliplatin-induced peripheral neuropathy using calmangafodipir (PledOx((R))): a placebo-controlled randomised phase II study (PLIANT). Acta Oncologica. 2018;57:393–402. [DOI] [PubMed] [Google Scholar]
- 155.Tournigand C, Cervantes A, Figer A, et al. OPTIMOX1: a randomized study of FOLFOX4 or FOLFOX7 with oxaliplatin in a stop-and-go fashion in advanced colorectal cancer—a GERCOR study. Journal of clinical oncology. 2006;24:394–400. [DOI] [PubMed] [Google Scholar]
- 156.Rao RD, Michalak JC, Sloan JA, et al. Efficacy of gabapentin in the management of chemotherapy-induced peripheral neuropathy. Cancer. 2007;110:2110–2118. [DOI] [PubMed] [Google Scholar]
- 157.Smith EML, Pang H, Cirrincione C, et al. Effect of duloxetine on pain, function, and quality of life among patients with chemotherapy-induced painful peripheral neuropathy: a randomized clinical trial. Jama. 2013;309:1359–1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Barton DL, Wos EJ, Qin R, et al. A double-blind, placebo-controlled trial of a topical treatment for chemotherapy-induced peripheral neuropathy: NCCTG trial N06CA. Supportive Care in Cancer. 2011;19:833–841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Rao RD, Flynn PJ, Sloan JA, et al. Efficacy of lamotrigine in the management of chemotherapy-induced peripheral neuropathy. Cancer. 2008;112:2802–2808. [DOI] [PubMed] [Google Scholar]