Abstract
Purpose of review
The purpose of this review is to provide a practical clinical approach to confusion in the patient with cancer. Confusion in the cancer population has a broader differential diagnosis than in the general medical population. The clinician must consider the usual differential diagnoses as well as causes unique to the cancer patient including direct complications from the cancer and indirect complications related to cancer treatment.
Recent findings
In the recent age of precision medicine, the oncologist now utilizes the genomic profile of both the patient and the tumor to provide advanced biologic therapies including targeted anticancer drugs, antiangiogenic agents, and immunotherapy. Such advances carry with them an emerging pattern of neurotoxicity which, although less well described in the literature, is now an important consideration to the clinical approach to confusion in cancer patients.
Summary
Confusion is the most common neurologic complication in cancer and is associated with significant morbidity, mortality, and prolonged hospital stays resulting in increased health care costs. Early recognition and treatment of delirium is essential to improve clinical outcomes.
Keywords: Encephalopathy in cancer, reversible delirium, precision medicine
Introduction
In 2016, the American Society of Clinical Oncology (ASCO) reported that 1.7 million Americans were diagnosed with cancer in the year 2015 and that this number will rise to 2.3 million in 2030.1,2 In this setting of increasing cancer rates, there has also been an explosion of new cancer therapies including: immune checkpoint inhibitors, T-cell therapies, tumor vaccines, antiangiogenic therapies, and various targeted agents, many of which cause frequent neurologic complications including confusion.3–7
Confusion in the cancer patient is common.8–11 Multiple synonymous terms for confusion exist, including: encephalopathy, delirium, acute confusional state, reversible dementia, organic brain syndrome, and terminal restlessness referring to the agitated delirium often seen in the last days of life.12 The incidence of confusion in cancer patients ranges from 57%–85 % compared to 15%–30% in medically ill hospitalized patients.13,14 In a prospective observational study of 352 cancer patients admitted to acute palliative care units (APCU) at the MD Anderson Cancer Center, delirium was the most common symptomatic complication occurring in 43% of patients at admission, and 70% during the entire APCU stay.15 In 2015, at Memorial Sloan Kettering Cancer Center (MSKCC), confusion or altered mental state accounted for 27% of all inpatient consultations requested of the Neurology service. This is a minimal incidence because it does not include the confused patients for whom a neurologic consultation was not called, those consulted by psychiatry, or those whose confusion was unrecognized.
Failure to recognize delirium in hospitalized patients is common, and may be due in part to the fluctuating nature of symptoms as well as failure to utilize proper tools for assessment and monitoring such as the Mini-Mental State Examination (MMSE) or Short Orientation Concentration Test (SOMCT). A retrospective review of 771 consecutive palliative care inpatient consults at a large cancer hospital found that 61% of delirium diagnoses were missed by the primary referring team and that hypoactive delirium was the most common subtype (63%).16 Given this high prevalence, cancer patients should be screened with an appropriate assessment on admission and throughout their hospital course.17 In the cancer patient hypoactive delirium due to hypoxia, infection, or metabolic encephalopathy is often missed and misinterpreted as fatigue, depression, or sedation from opioid pain medication. Cancer predominately affects individuals age 65 and older, and older age and a pre-existing psychiatric condition are associated with a missed diagnosis of delirium which is often misinterpreted as dementia in this population.16,18,19
Delirium is associated with increased morbidity and mortality, prolonged hospital and intensive care unit (ICU) stays, as well as increased distress in patients, families, and caregivers.20 To improve outcomes, the clinician must have a rational approach to the confused cancer patient, and Inouye’s model of delirium is a good starting point.21 The clinician must recognize the predisposing risk factors for confusion (such as old age, dementia, mood disorder, substance abuse, hypoalbuminemia, cachexia, advanced cancer, bone metastases) and determine the more immediate precipitating factors which may be modifiable. Delirium is reversible in approximately 50% of cancer patients, and may require correction of only some and not all contributing causes.13 Frequently, multiple causes, a median of three in one series, are identified in patients with cancer.8 Confusion attributed to infection, electrolyte abnormalities, or medications is more likely reversible than when the delirium is the consequence of global hypoxic encephalopathy. The additional considerations in the confused cancer patient are those factors directly related to the cancer and its treatment.
Confusion: Direct Relation to Cancer
The differential diagnosis of confusion directly related to cancer is extensive and includes metabolic abnormalities, metastatic disease, vascular disorders, paraneoplastic and autoimmune syndromes, and inflammatory mediators (Table 1). The etiology is typically multifactorial. When a single cause for confusion is identified, it is usually structural such as multiple brain metastases, meningitis/encephalitis, or cerebral infarction. The clinical evaluation should include a careful history to identify pre-existing risk factors and new interventions such as medications (Table 2). The neurologic examination may indicate lateralizing neurologic signs suggestive of brain metastases, signs of raised intracranial pressure such as papilledema, abducens nerve palsy indicating leptomeningeal metastasis (LM), or stupor from a global hypoxic-ischemic or metabolic encephalopathy. A complete laboratory evaluation is needed to assess for metabolic and immune conditions unique to cancer patients including tumor lysis syndrome (TLS), hypercalcemia of malignancy, cachexia/anorexia and paraneoplastic autoimmune syndromes (PNS/AIS).
Table 1.
Etiologies of Confusion: Cancer Related
Metabolic | ||
Electrolyte abnormality (hypomagnesemia, hypophosphatemia, hypo- hypernatremia) | ||
Tumor lysis syndrome | ||
Hypercalcemia of malignancy | ||
Cachexia/anorexia syndrome | ||
Thiamine deficiency | ||
Carcinoid syndrome/pellagra | ||
Endocrine dysfunction | ||
Adrenal-Addison/Cushing disease | ||
Thyroid-myxedema, thyrotoxicosis | ||
Parathyroid-hypo- and hyperparathyroidism | ||
End organ failure | ||
Liver-hepatic encephalopathy | ||
Kidney-uremic encephalopathy | ||
Brain-hypoxic encephalopathy | ||
Lung-carbon monoxide narcosis | ||
Metastatic | ||
Brain | ||
Leptomeningeal | ||
Gliomatosis/Lymphomatosis | ||
Vascular | ||
Embolic | ||
Nonbacterial thrombotic endocarditis | ||
Tumor (myxoma, lung, mucin secreting) | ||
Bone marrow | ||
Fat | ||
Infarction | ||
Disseminated intravascular coagulation (leukemia, breast, lung) | ||
Hypercoagulation | ||
Thrombocytosis (myeloproliferative disorders) | ||
Leukostasis (leukemia) | ||
Hyperviscosity (leukemia, multiple myeloma) | ||
Arterial and venous occlusion by direct tumor invasion | ||
Hemorrhage | ||
Hypocoagulation (myeloproliferative disorders) | ||
Brain metastases (melanoma, lung, germ cell, kidney) | ||
Thrombotic microangiopathy (gastric, breast, lung) | ||
Vasculitis (Hairy cell leukemia, Hodgkin disease) | ||
Intravascular lymphoma/Angiotropic lymphoma | ||
Limbic encephalitis | ||
Cancer | Associated antibody | |
Paraneoplastic | Small cell lung | Anti-Hu |
Germ cell, testes | Anti-Ma | |
Hodgkin lymphoma (Ophelia syndrome) |
NMDAR, mGluR5, Hu | |
Autoimmune | Ovarian teratoma | NMDA |
Breast | AMPA | |
Neuroendocrine lung | GABA b | |
Thymus | LGI1 | |
Inflammatory | ||
Cytokine release | ||
Interleukin 1, 6, and 8, interferon, tumor necrosis factor |
Abbreviations: NMDAR – N-methyl-D-aspertate receptor; mGluR5 – Metabotropic glutamate receptor 5; AMPA – Alpha-amino-3-hydroxy-5-methyl- isoxazolepropionic acid receptor; GABA b – gamma-aminobutyric acid; LGI1 – Leucine-rich, glioma inactivated 1
Table 2.
Clinical Evaluation of the Confused Cancer Patient
Risk factors |
Advanced age |
Advanced cancer |
Pre-existing cognitive dysfunction |
Pre-existing psychiatric disorder |
Cardiovascular disease |
Diabetes |
Substance abuse |
Neurologic examination |
State of consciousness |
Pupillary abnormalities, eye movements, funduscopic exam |
Asterixis |
Myoclonus |
Muscle tone |
Focal signs (hemiparesis, aphasia, ataxia) |
Laboratory testing |
Structural |
Brain MRI with gadolinium: metastases, hemorrhage, infarction, infection |
Lumbar puncture: leptomeningeal disease, increased intracranial pressure, infection |
EEG: nonconvulsive status epilepticus, triphasic waves |
Blood |
Complete blood count |
Electrolytes (Na, K, Ca, Mg, Po4) |
BUN and creatinine |
Liver function and ammonia |
Glucose |
Thyroid function and cortisol |
Lactate |
Arterial blood gas |
Thiamine, folic acid, Vitamin B12 |
Blood cultures |
Autoimmune antibodies |
Brain Metastases
Brain metastases (BM) occur in 10–30% of cancer patients and are most common in lung and breast carcinoma followed by melanoma.22 In a retrospective review of inpatients with BM over a 6 month period, 45.8% had cognitive changes.23 Cognitive impairment is the initial complaint in approximately 30% of patients with BM, and often occurs with lateralizing signs. However, multiple small BM may cause confusion and personality change only. Some focal abnormalities such as an apraxia, aphasia, and abulia from metastases in the parietal or frontal lobes may be mistaken for confusion.
Worsening confusion in association with headache and disorientation is concerning for increased intracranial pressure potentially from vasogenic edema associated with BM or hydrocephalus from LM. In this setting, episodes of depressed consciousness, speech arrest, or loss of leg tone may occur and most likely represent plateau waves rather than seizure.
Tumor Lysis Sydrome
TLS occurs as the result of treatment-induced tumor cell death releasing intracellular potassium, phosphorus, and uric acid, causing hypocalcemia, hyperkalemia, and precipitating renal insufficiency or failure. 24–27 Volume depletion and hematologic cancers associated with a high tumor burden are the major risk factors. Encephalopathy and seizures are common symptoms; prevention and treatment require aggressive hydration, urine alkalinization, and allopurinol or rasburicase to reduce urate levels.
Hypercalcemia of Malignancy
Hypercalcemia of malignancy occurs in 20% of patients with cancer, most commonly associated with multiple myeloma (MM), leukemia and non-Hodgkin lymphoma28,29 and among solid tumors, breast, lung, and renal cancer. Approximately 80% of cases are humoral, associated with parathyroid hormone-related protein (PTHrP) which increases bone resorption through osteoclast activation and osteoblast suppression.30,31 Less common causes are direct metastatic involvement of bone and 1,25 dihydroxy vitamin D(calcitriol) and parathyroid hormone- mediated hypercalcemia.32 A rapid rise of serum calcium (>13.5 mg/dL) causes delirium and coma. Chronic elevation may produce more subtle findings such as malaise, lethargy, mood disturbance, anorexia, and constipation, constitutional symptoms which may mistakenly be attributed to the patient’s underlying disease. Hydration and bisphosphonates are the main treatments. Denosumab, a monoclonal antibody that inhibits osteoclast function, is useful for bisphosphonate-refractory hypercalcemia.33
Cachexia, Anorexia, and Malnutrition
Nutritional deficiencies and cachexia, which is mediated by chronic inflammation, have been associated with cognitive decline.34–37 The tumor, and its humoral response produce inflammatory cytokines, particularly IL-1, IL-6, and tumor necrosis factor alpha (TNF-a) which are associated with delirium and cognitive dysfunction by affecting serotonin and hypothalamic pathways.38–40
Disorders due to deprivation of a single nutrient have been reported in cancer patients including the Wernicke Korsakoff syndrome (WKS) from thiamine deficiency, vitamin B12 deficiency, and niacin deficiency associated with carcinoid tumors; the latter two are uncommon. WKS is characterized by cognitive impairment, ocular signs, and ataxia and has traditionally been described with alcohol use, but is seen in the oncologic population.41 In a large retrospective review of WKS in cancer patients, the most common sign was cognitive impairment (100%) followed by ataxia (39%) and oculomotor signs (17%); MRI findings typical of WKS were found in only 30% of patients.42 Thiamine deficiency may result from malabsorption due to a structural lesion or secondary to diminished appetite, diarrhea, or vomiting. It may also result from hypermetabolism associated with illness from sepsis, fever, and steroid use. Rapidly growing tumors and hematologic malignancies can deplete thiamine stores due to rapid cellular turn over. Fluorouracil, a drug used to treat gastric and head and neck cancers, can produce thiamine deficiency.43 Thiamine supplementation should be given to any cancer patient presenting with confusion.
PNS/AIS
Immune-mediated disorders, including classic paraneoplastic neurologic syndromes and more recently described autoimmune syndromes (AIS), can cause limbic encephalitis (LE) (Table 1).44–47 Unlike classic PNS, the AIS occur more commonly in younger patients, with or without a cancer diagnosis, and are responsive to treatment; the associated antibodies target extracellular surface antigens as opposed to intracellular neuronal antigens in PNS.
Antibodies most frequently associated with LE are Hu (small cell lung) and Ma (testicular) in PNS. LE is characterized by confusion, memory impairment, and psychiatric symptoms in association with CSF findings of pleocytosis, elevated IgG, or oligoclonal bands. MRI may show bilateral abnormalities in the medial temporal lobe on T2 weighted fluid- attenuated inversion recovery (FLAIR) imaging. Antibodies in the CSF are not necessary to establish the diagnosis, however, testing for these antibodies is important because their presence may indicate a specific tumor type as well as predict outcome. LE from AIS has a better response to immunotherapy and anti-tumor treatment and a better prognosis with a recovery of 80%.
Confusion: Indirect Relation to Cancer Treatment
Confusion attributed to cancer therapy should be a diagnosis of exclusion after considering cancer related causes, particularly metastatic disease. Many cancer treatments can cause confusion (Table 3), and this review will focus upon those associated with acute encephalopathy, posterior reversible encephalopathy syndrome (PRES), and progressive multifocal leukoencephalopathy (PML) (Table 4).
Table 3.
Confusion: Cancer Treatment Related
I. |
Cancer medications a) Supportive medications Antihistamines Amphetamines Anticholinergics Appetite stimulants (mitotane, aminoglutethimide) Benzodiazepines Corticosteroids H2 blockers Opioids b) Antineoplastic agents Standard chemotherapy agents Intrathecal chemotherapy Small –molecule inhibitors (PRES) Anti-angiogenic agents (stroke/PRES) Immune therapy (encephalopathy/PRES/PML) Targeted B and T cell agents CAR T –cell transfusion Immune check point inhibitors |
II. |
Hematopoietic stem cell transplantation Induction therapy (seizure /encephalopathy) Chronic immunosuppression (opportunistic infection, PML, PRES) |
III. |
Cranial radiation Radiation necrosis Delayed leukoencephalopathy |
IV. |
Surgery Pre-operative (old age, dementia, alcoholism, cardiac disease, diabetes) Intra-operative (hypoperfusion and anoxia, cerebral emboli) Post-operative (insomnia, medications, nutritional abnormality, infection, pulmonary embolus) |
Abbreviations: PRES - posterior reversible encephalopathy syndrome; PML - progressive multifocal leukoencephalopathy
Table 4.
Specific Cancer Therapeutics That Cause Confusion
Encephalopathy | Seizure | PRES | PML |
---|---|---|---|
Acute | Busulfan | Bevacizumab | Alemtuzumab |
Bortezomib | CAR T-cell therapy | Blinatumomab | Brentuximab |
Blinatumomab | Carmustine | Bortezomib | Fludarabine |
CAR T-cell therapy | Cisplatin | CAR T-cell therapy | Rituximab |
Cisplatin | Cytarabine | Cisplatin | Rutolitinib |
Etoposide | Etoposide | Carboplatin | |
Fludarabine | Fludarabine | Docetaxel | |
5-Fluorouracil | Ifosfamide | Ipilimumab | |
Ifosfamide | L-Asparaginase | Irinotecan | |
Interferon-alpha | Methotrexate | Oxaliplatin | |
Interleukin-1 | Vincristine | Paclitaxel | |
Ipilimumab | Pazopanib | ||
L-Asparaginase | Rituximab | ||
Methotrexate | Sirolimus | ||
Nitrosoureas | Sorafenib | ||
Procarbazine | Sunitinib | ||
Tamoxifen | Tacrolimus | ||
Thiotepa | Vincristine | ||
Vincristine | |||
Chronic | |||
Carmustine | |||
Cytarabine | |||
Fludarabine | |||
Methotrexate |
Abbreviations: PRES – Posterior reversible encephalopathy syndrome; PML – Progressive multifocal encephalopathy
Acute confusion may occur after the use of antineoplastic agents that cross the blood brain barrier (BBB) such as high dose cytarabine or methotrexate, procarbazine, 5-flourouracil, and the nitrosoureas.48,49 Most episodes are idiosyncratic and reversible after drug withdrawal. However, ifosfamide encephalopathy can occur hours to days after administration and may be fatal; methylene blue may be helpful.50 High dose methotrexate may produce a unique stroke-like syndrome of alternating focal deficits with encephalopathy, usually transient, and occurring several days after the second or third cycle.51 Other than an idiosyncratic reaction, confusion may be the result of an electrolyte disorder particularly with platins that cause hypomagnesemia, hypokalemia, and hypocalcemia, and the vinca alkaloids which may cause hyponatremia from SIADH.52 Seizure can manifest as encephalopathy resulting from either an electrolyte disturbance or from drug itself such as high dose busulphan.53,54 Partial complex seizures may present only as confusion and can be prolonged from non-convulsive status epilepticus which is detectable only by EEG. Seizures may also occur with intrathecal administration of any drug including methotrexate, cytarabine, thiotepa, steroids, or rituximab.
CNS complications occur in 10–65% of patients who undergo hematopoietic stem cell transplantation (HSCT) and are associated with poor outcome.55,56 Most neurologic complications occur within 100 days of transplant and manifest as seizure and encephalopathy. Seizures are most common immediately after the stem cell infusion and may be related to dimethyl sulfoxide used for cryopreservation. High dose busulphan and total body irradiation are other contributing factors during induction. Pancytopenia prior to bone marrow reconstitution and long term immunosuppression after allotransplantation predispose patients to delayed opportunistic CNS infections.
Small molecule inhibitors and novel T-cell therapies may cause confusion. Bortezomib, a proteasome inhibitor, has been associated with acute change in mental status.57 Blinatumomab is a bispecific T-cell engaging antibody that is effective in acute lymphocytic leukemia.58,59 Somnolence, seizure, and encephalopathy occur in 15%–20% of treated patients. CNS symptoms typically resolve with discontinuation of the drug and have been attributed to acute release of inflammatory cytokines. A similar syndrome is seen after CAR T -cell therapy which is used to treat lymphoma or leukemia.60–63 It is often associated with confusion, psychiatric symptoms, seizure, and encephalopathy occurring within weeks of T-cell infusion. The etiology of the neurologic symptoms may be direct T-cell toxicity in the brain as CAR T-cells cross the BBB and are found in the CSF, or through systemic inflammatory mediators producing a cytokine release syndrome.64 The cytokine release syndrome can range clinically from a mild flu-like syndrome with fever to multisystem organ failure and severe encephalopathy.
Ipilimumab, an immune check point inhibitor, is associated with encephalopathy.65 Pathogenesis is thought to be due to elevated levels of proinflammatory cytokines. Ipilimumab and other check point inhibitors such as nivolumab and pembrolizumab may be associated with an autoimmune thyroiditis which can present with confusion66–68. In addition, autoimmune hypophysitis with confusion and diabetes insipidus is seen in 3% of patients treated with these agents. One case of ipilimumab-induced encephalopathy with a reversible splenial lesion in the corpus callosum has been reported.69 The full spectrum of CNS toxicities from immune checkpoint inhibitors likely remains to be defined.
Posterior reversible encephalopathy syndrome (PRES) is a well-characterized clinico-radiographic entity of altered mental status and seizure in association with characteristic neuroimaging findings in the posterior white matter. In two large reviews of PRES in cancer patients, confusion occurred in 59%–71% of patients.70,71 PRES is associated with hypertension, the puerperium, and immunosuppressive agents used in organ and HSCT; more recently, it has been described with targeted agents, anti-angiogenic agents, and immune check point inhibitors.72–75
In one review of 31 cancer patients, 55% had received chemotherapy or targeted agents in the month preceding PRES, with 35% receiving bevacizumab.70 PRES typically resolves when the causative drug is withheld, but residual deficits from infarction or hemorrhage are more common in cancer patients.76
PML, a fatal demyelinating disease caused by activation of the latent John Cunningham (JC) virus in an immunosuppressed patient, is a rare cause of confusion in cancer patients. PML is increasingly seen in those with hematologic cancers treated with monoclonal antibodies that target B and T cells such as rituximab, brentuximab, and alemtuzumab, and small molecules such as ruxolitinib.77–79 In addition, it is increasingly recognized as a complication of allogeneic HSCT due to chronic immunosuppression needed to prevent graft versus host disease.80 Although brain biopsy is the gold standard for diagnosis, detection of JC virus in the CSF in association with typical white matter changes on neuroimaging is sufficient to confirm the diagnosis; however, the sensitivity of CSF PCR for JC virus is only 75% so in patients where PML is considered likely, a brain biopsy may still be necessary for diagnosis.77,81 Reconstitution of immune function is the only known treatment.
Conclusion
A rational clinical approach to the confused oncologic patient is one which identifies both direct and indirect related etiologies. Cancer itself affects the normal biologic milieu resulting in inflammatory, metabolic, and neurochemical changes not present in the general medical population. Such changes precipitate confusion from both resultant increased BBB permeability and systemic end organ dysfunction. With the rapid development of “mutation-based” clinical trials of targeted therapeutics and the growing armamentarium of biologic therapies, it is essential for the clinician to be familiar with the increasing association of delirium to newer agents.
Key Points.
Confusion is the most common neurologic symptom in cancer and may be directly related to cancer itself or to cancer treatment.
The under recognition of potentially reversible causes for delirium in the cancer patient results in prolonged hospital stays, distress for patient, family, and caregivers, and increased morbidity and mortality.
The current era of precision medicine is rapidly producing novel targeted and immune therapeutics that are associated with an emerging pattern of neurotoxicity that includes confusion as a common manifestation.
Acknowledgments
We would like to thank Judith Lampron for her help with the editing of this manuscript.
Financial support and sponsorship: This research was funded in part through the NIH/NCI Cancer Center Support Grant P30 CA008748.
Footnotes
The authors have no conflict of interest
This manuscript has been seen, reviewed and approved by all contributing authors
References
- 1.Society AC. American Cancer Society Facts & Figures 2015. 2015 2015: http/www.cancerorg/acs/groups/content/@editorial/documents/document/acspc-044552.pdf.
- 2.Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–2921. doi: 10.1158/0008-5472.CAN-14-0155. [DOI] [PubMed] [Google Scholar]
- 3**.Dizon DS, Krilov L, Cohen E, et al. Clinical Cancer Advances 2016: Annual Report on Progress Against Cancer From the American Society of Clinical Oncology. J Clin Oncol. 2016;34(9):987–1011. doi: 10.1200/JCO.2015.65.8427. The clinician evaluating a confused cancer patient should be familiar with the recent advances in cancer therapeutics as reported by ASCO in this report. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Soffietti R, Trevisan E, Ruda R. Neurologic complications of chemotherapy and other newer and experimental approaches. Handb Clin Neurol. 2014;121:1199–1218. doi: 10.1016/B978-0-7020-4088-7.00080-8. [DOI] [PubMed] [Google Scholar]
- 5.Magge RS, DeAngelis LM. The double-edged sword: Neurotoxicity of chemotherapy. Blood Rev. 2015;29(2):93–100. doi: 10.1016/j.blre.2014.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Nolan CP, DeAngelis LM. Neurologic Complications of Chemotherapy and Radiation. In: Continuum A, editor. Neuro-Oncology. Minneapolis: 2015. pp. 429–451. [DOI] [PubMed] [Google Scholar]
- 7*.Stone JB, DeAngelis LM. Cancer-treatment-induced neurotoxicity–focus on newer treatments. Nat Rev Clin Oncol. 2016;13(2):92–105. doi: 10.1038/nrclinonc.2015.152. This is the most recent review of the neurotoxic effects of cancer treatment with a particular focus on the newer therapeutics, such as targeted anticancer agents and immunotherapy. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Tuma R, DeAngelis LM. Altered mental status in patients with cancer. Arch Neurol. 2000;57(12):1727–1731. doi: 10.1001/archneur.57.12.1727. [DOI] [PubMed] [Google Scholar]
- 9*.Lawlor PG, Bush SH. Delirium in patients with cancer: assessment, impact, mechanisms and management. Nat Rev Clin Oncol. 2015;12(2):77–92. doi: 10.1038/nrclinonc.2014.147. A comprehensive review of the assessment tools, the potential pathogenesis, and the medical management of delirium. The review highlights the distress that delirium often brings to patients, families, and care givers. [DOI] [PubMed] [Google Scholar]
- 10**.de la Cruz M, Ransing V, Yennu S, et al. The Frequency, Characteristics, and Outcomes Among Cancer Patients With Delirium Admitted to an Acute Palliative Care Unit. Oncologist. 2015;20(12):1425–1431. doi: 10.1634/theoncologist.2015-0115. This large retrospective review of cancer patients finds that delirium is very common in cancer patients and is associated with lower overall survival. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Williams AM, Janelsins MC, van Wijngaarden E. Cognitive function in cancer survivors: analysis of the 1999–2002 National Health and Nutrition Examination Survey. Support Care Cancer. 2016;24(5):2155–2162. doi: 10.1007/s00520-015-2992-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Bush SH, Leonard MM, Agar M, et al. End-of-life delirium: issues regarding recognition, optimal management, and the role of sedation in the dying phase. J Pain Symptom Manage. 2014;48(2):215–230. doi: 10.1016/j.jpainsymman.2014.05.009. [DOI] [PubMed] [Google Scholar]
- 13.Boettger S, Jenewein J, Breitbart W. Delirium and severe illness: Etiologies, severity of delirium and phenomenological differences. Palliat Support Care. 2015;13(4):1087–1092. doi: 10.1017/S1478951514001060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Grassi L, Caraceni A, Mitchell AJ, et al. Management of delirium in palliative care: a review. Curr Psychiatry Rep. 2015;17(3):550. doi: 10.1007/s11920-015-0550-8. [DOI] [PubMed] [Google Scholar]
- 15*.Hui D, dos Santos R, Reddy S, et al. Acute symptomatic complications among patients with advanced cancer admitted to acute palliative care units: A prospective observational study. Palliat Med. 2015;29(9):826–833. doi: 10.1177/0269216315583031. This is the first prospective study to document delirium as the most common complication at admission to a large inpatient palliative care unit. [DOI] [PubMed] [Google Scholar]
- 16**.de la Cruz M, Fan J, Yennu S, et al. The frequency of missed delirium in patients referred to palliative care in a comprehensive cancer center. Support Care Cancer. 2015;23(8):2427–2433. doi: 10.1007/s00520-015-2610-3. This large retrospective review reports the high incidence of missed delirium among cancer patients, most often associated with hypoactive delirium. The study reminds the clinician not to misinterpret the later with mood disorder, dementia, or opioid use. [DOI] [PubMed] [Google Scholar]
- 17.Uchida M, Okuyama T, Ito Y, et al. Prevalence, course and factors associated with delirium in elderly patients with advanced cancer: a longitudinal observational study. Jpn J Clin Oncol. 2015;45(10):934–940. doi: 10.1093/jjco/hyv100. [DOI] [PubMed] [Google Scholar]
- 18.Korc-Grodzicki B, Holmes HM, Shahrokni A. Geriatric assessment for oncologists. Cancer Biol Med. 2015;12(4):261–274. doi: 10.7497/j.issn.2095-3941.2015.0082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mandelblatt JS, Jacobsen PB, Ahles T. Cognitive effects of cancer systemic therapy: implications for the care of older patients and survivors. J Clin Oncol. 2014;32(24):2617–2626. doi: 10.1200/JCO.2014.55.1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Serafim RB, Bozza FA, Soares M, et al. Pharmacologic prevention and treatment of delirium in intensive care patients: A systematic review. J Crit Care. 2015;30(4):799–807. doi: 10.1016/j.jcrc.2015.04.005. [DOI] [PubMed] [Google Scholar]
- 21.Inouye SK, Charpentier PA. Precipitating factors for delirium in hospitalized elderly persons. Predictive model and interrelationship with baseline vulnerability. JAMA. 1996;275(11):852–857. [PubMed] [Google Scholar]
- 22.Lin X, DeAngelis LM. Treatment of Brain Metastases. J Clin Oncol. 2015;33(30):3475–3484. doi: 10.1200/JCO.2015.60.9503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Gofton TE, Graber J, Carver A. Identifying the palliative care needs of patients living with cerebral tumors and metastases: a retrospective analysis. J Neurooncol. 2012;108(3):527–534. doi: 10.1007/s11060-012-0855-y. [DOI] [PubMed] [Google Scholar]
- 24.Howard SC, Jones DP, Pui CH. The tumor lysis syndrome. N Engl J Med. 2011;364(19):1844–1854. doi: 10.1056/NEJMra0904569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zivin SP, Elias Y, Ray CE., Jr Tumor lysis syndrome and primary hepatic malignancy: case presentation and review of the literature. Semin Intervent Radiol. 2015;32(1):3–9. doi: 10.1055/s-0034-1396956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mirrakhimov AE, Voore P, Khan M, Ali AM. Tumor lysis syndrome: A clinical review. World J Crit Care Med. 2015;4(2):130–138. doi: 10.5492/wjccm.v4.i2.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27*.Howard SC, Trifilio S, Gregory TK, Baxter N, McBride A. Tumor lysis syndrome in the era of novel and targeted agents in patients with hematologic malignancies: a systematic review. Ann Hematol. 2016;95(4):563–573. doi: 10.1007/s00277-015-2585-7. This article reviews phase I-III clinical trials of new targeted agents that may be associated with TLS. [DOI] [PubMed] [Google Scholar]
- 28.Jick S, Li L, Gastanaga VM, Liede A. Prevalence of hypercalcemia of malignancy among cancer patients in the UK: analysis of the Clinical Practice Research Datalink database. Cancer Epidemiol. 2015;39(6):901–907. doi: 10.1016/j.canep.2015.10.012. [DOI] [PubMed] [Google Scholar]
- 29.Goldner W. Cancer-Related Hypercalcemia. J Oncol Pract. 2016;12(5):426–432. doi: 10.1200/JOP.2016.011155. [DOI] [PubMed] [Google Scholar]
- 30*.Sternlicht H, Glezerman IG. Hypercalcemia of malignancy and new treatment options. Ther Clin Risk Manag. 2015;11:1779–1788. doi: 10.2147/TCRM.S83681. A comprehensive review of the etiology, clinical presentation, pathogenesis, and treatment of hypercalcemia of malignancy. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Galindo RJ, Romao I, Valsamis A, Weinerman S, Harris YT. Hypercalcemia of Malignancy and Colorectal Cancer. World J Oncol. 2016;7(1):5–12. doi: 10.14740/wjon953w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mirrakhimov AE. Hypercalcemia of Malignancy: An Update on Pathogenesis and Management. N Am J Med Sci. 2015;7(11):483–493. doi: 10.4103/1947-2714.170600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Castellano D, Sepulveda JM, Garcia-Escobar I, Rodriguez-Antolin A, Sundlov A, Cortes-Funes H. The role of RANK-ligand inhibition in cancer: the story of denosumab. Oncologist. 2011;16(2):136–145. doi: 10.1634/theoncologist.2010-0154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Sanders C, Behrens S, Schwartz S, et al. Nutritional Status is Associated with Faster Cognitive Decline and Worse Functional Impairment in the Progression of Dementia: The Cache County Dementia Progression Study1. J Alzheimers Dis. 2016;52(1):33–42. doi: 10.3233/JAD-150528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Smith PJ, Blumenthal JA. Dietary Factors and Cognitive Decline. J Prev Alzheimers Dis. 2016;3(1):53–64. doi: 10.14283/jpad.2015.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Porporato PE. Understanding cachexia as a cancer metabolism syndrome. Oncogenesis. 2016;5:e200. doi: 10.1038/oncsis.2016.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Molfino A, Gioia G, Rossi Fanelli F, Laviano A. Contribution of Neuroinflammation to the Pathogenesis of Cancer Cachexia. Mediators Inflamm. 2015;2015:801685. doi: 10.1155/2015/801685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ritter C, Tomasi CD, Dal-Pizzol F, et al. Inflammation biomarkers and delirium in critically ill patients. Crit Care. 2014;18(3):R106. doi: 10.1186/cc13887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Brum C, Stertz L, Borba E, Rumi D, Kapczinski F, Camozzato A. Association of serum brain-derived neurotrophic factor (BDNF) and tumor necrosis factor-alpha (TNF-alpha) with diagnosis of delirium in oncology inpatients. Rev Bras Psiquiatr. 2015;37(3):197–202. doi: 10.1590/1516-4446-2014-1450. [DOI] [PubMed] [Google Scholar]
- 40.Vasunilashorn SM, Ngo L, Inouye SK, et al. Cytokines and Postoperative Delirium in Older Patients Undergoing Major Elective Surgery. J Gerontol A Biol Sci Med Sci. 2015;70(10):1289–1295. doi: 10.1093/gerona/glv083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Scalzo SJ, Bowden SC, Ambrose ML, Whelan G, Cook MJ. Wernicke-Korsakoff syndrome not related to alcohol use: a systematic review. J Neurol Neurosurg Psychiatry. 2015;86(12):1362–1368. doi: 10.1136/jnnp-2014-309598. [DOI] [PubMed] [Google Scholar]
- 42**.Isenberg-Grzeda E, Alici Y, Hatzoglou V, Nelson C, Breitbart W. Nonalcoholic Thiamine-Related Encephalopathy (Wernicke-Korsakoff Syndrome) Among Inpatients With Cancer: A Series of 18 Cases. Psychosomatics. 2016;57(1):71–81. doi: 10.1016/j.psym.2015.10.001. This is the largest series of cancer patients with WKS reported to date. The study discusses mechanisms of thiamine deficiency unique to cancer patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Cho KP, Lee JS, Seong JS, et al. Two cases of Wernicke s encephalopathy that developed during total parenteral nutrition in colon cancer patients treated with 5-fluorouracil-based chemotherapy. Korean J Gastroenterol. 2014;64(3):158–163. doi: 10.4166/kjg.2014.64.3.158. [DOI] [PubMed] [Google Scholar]
- 44.Dalmau J, Rosenfeld MR. Autoimmune encephalitis update. Neuro Oncol. 2014;16(6):771–778. doi: 10.1093/neuonc/nou030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Joubert B, Kerschen P, Zekeridou A, et al. Clinical Spectrum of Encephalitis Associated With Antibodies Against the alpha-Amino-3-Hydroxy-5-Methyl-4-Isoxazolepropionic Acid Receptor: Case Series and Review of the Literature. JAMA Neurol. 2015;72(10):1163–1169. doi: 10.1001/jamaneurol.2015.1715. [DOI] [PubMed] [Google Scholar]
- 46.Moreira DM, Gershman B, Lohse CM, et al. Paraneoplastic syndromes are associated with adverse prognosis among patients with renal cell carcinoma undergoing nephrectomy. World J Urol. 2016 doi: 10.1007/s00345-016-1793-7. [DOI] [PubMed] [Google Scholar]
- 47*.Graus F, Titulaer MJ, Balu R, et al. A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol. 2016;15(4):391–404. doi: 10.1016/S1474-4422(15)00401-9. The review article provides clinical guidelines to the diagnostic approach to autoimmune encephalitis and discusses the prognostic role of onconeuronal antibodies. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Matsuoka H, Yoshiuchi K, Koyama A, Otsuka M, Nakagawa K. Chemotherapeutic drugs that penetrate the blood-brain barrier affect the development of hyperactive delirium in cancer patients. Palliat Support Care. 2015;13(4):859–864. doi: 10.1017/S1478951514000765. [DOI] [PubMed] [Google Scholar]
- 49.Thomas SA, Tomeh N, Theard S. Fluorouracil-induced Hyperammonemia in a Patient with Colorectal Cancer. Anticancer Res. 2015;35(12):6761–6763. [PubMed] [Google Scholar]
- 50.Yeo KK. The use of continuous veno-venous hemodiafiltration in the management of ifosfamide-induced encephalopathy: A case report. J Pediatr Hematol Oncol. 2016 doi: 10.1097/MPH.0000000000000527. [DOI] [PubMed] [Google Scholar]
- 51.Gosavi TD, Ahmad MT, Lee LH, Lim SH. Methotrexate induced leucoencephalopathy: A stroke mimic. Ann Indian Acad Neurol. 2013;16(3):418–421. doi: 10.4103/0972-2327.116922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Liamis G, Filippatos TD, Elisaf MS. Electrolyte disorders associated with the use of anticancer drugs. Eur J Pharmacol. 2016;777:78–87. doi: 10.1016/j.ejphar.2016.02.064. [DOI] [PubMed] [Google Scholar]
- 53.Floeter AE, McCune JS. Levetiracetam for the prevention of busulfan-induced seizures in pediatric hematopoietic cell transplantation recipients. J Oncol Pharm Pract. 2016 doi: 10.1177/1078155216651128. [DOI] [PubMed] [Google Scholar]
- 54.Pasquini MC, Le-Rademacher J, Zhu X, et al. Intravenous Busulfan-Based Myeloablative Conditioning Regimens Prior to Hematopoietic Cell Transplantation for Hematologic Malignancies. Biol Blood Marrow Transplant. 2016 doi: 10.1016/j.bbmt.2016.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kang JM, Kim YJ, Kim JY, et al. Neurologic complications after allogeneic hematopoietic stem cell transplantation in children: analysis of prognostic factors. Biol Blood Marrow Transplant. 2015;21(6):1091–1098. doi: 10.1016/j.bbmt.2015.02.007. [DOI] [PubMed] [Google Scholar]
- 56.Aki SZ, Pamukcuoglu M, Bagriacik U, Sucas GT. Encephalopathy: an unusual neurologic complication of autologous hematopoietic stem cell transplant in patients with multiple myeloma with renal failure. Leuk Lymphoma. 2013;54(4):894–896. doi: 10.3109/10428194.2012.721545. [DOI] [PubMed] [Google Scholar]
- 57.Lao CD, Friedman J, Tsien CI, et al. Concurrent whole brain radiotherapy and bortezomib for brain metastasis. Radiat Oncol. 2013;8:204. doi: 10.1186/1748-717X-8-204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Jabbour E, O’Brien S, Ravandi F, Kantarjian H. Monoclonal antibodies in acute lymphoblastic leukemia. Blood. 2015;125(26):4010–4016. doi: 10.1182/blood-2014-08-596403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59*.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: 10.1182/blood-2015-06-651380. In this phase 2 study, blinatumomab monotherapy for treatment of refractory diffuse large B –cell lymphoma was associated with grade 3 encephalopathy. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224) doi: 10.1126/scitranslmed.3008226. 224ra225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Porter DL, Hwang WT, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303r. doi: 10.1126/scitranslmed.aac5415. a139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Maude SL, Teachey DT, Porter DL, Grupp SA. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood. 2015;125(26):4017–4023. doi: 10.1182/blood-2014-12-580068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63**.Davila ML, Sauter C, Brentjens R. CD19-Targeted T Cells for Hematologic Malignancies: Clinical Experience to Date. Cancer J. 2015;21(6):470–474. doi: 10.1097/PPO.0000000000000153. This article reviews the neurologic toxicities associated with CAR T cell infusion that have been reported in clinical trials, particularly CRS, and discusses management of these toxicities. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64*.Teachey DT, Lacey SF, Shaw PA, et al. Identification of Predictive Biomarkers for Cytokine Release Syndrome after Chimeric Antigen Receptor T-cell Therapy for Acute Lymphoblastic Leukemia. Cancer Discov. 2016;6(6):664–679. doi: 10.1158/2159-8290.CD-16-0040. The authors of this study measure serum cytokines in CTLO-19-treated patients to develop clinical biomarkers that may predict who will develop cytokine release syndrome. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Johnson DB, Friedman DL, Berry E, et al. Survivorship in Immune Therapy: Assessing Chronic Immune Toxicities, Health Outcomes, and Functional Status among Long-term Ipilimumab Survivors at a Single Referral Center. Cancer Immunol Res. 2015;3(5):464–469. doi: 10.1158/2326-6066.CIR-14-0217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ryder M, Callahan M, Postow MA, Wolchok J, Fagin JA. Endocrine-related adverse events following ipilimumab in patients with advanced melanoma: a comprehensive retrospective review from a single institution. Endocr Relat Cancer. 2014;21(2):371–381. doi: 10.1530/ERC-13-0499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Faje AT, Sullivan R, Lawrence D, et al. Ipilimumab-induced hypophysitis: a detailed longitudinal analysis in a large cohort of patients with metastatic melanoma. J Clin Endocrinol Metab. 2014;99(11):4078–4085. doi: 10.1210/jc.2014-2306. [DOI] [PubMed] [Google Scholar]
- 68.Carl D, Grullich C, Hering S, Schabet M. Steroid responsive encephalopathy associated with autoimmune thyroiditis following ipilimumab therapy: a case report. BMC Res Notes. 2015;8:316. doi: 10.1186/s13104-015-1283-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Conry RM, Sullivan JC, Nabors LB., 3rd Ipilimumab-induced encephalopathy with a reversible splenial lesion. Cancer Immunol Res. 2015;3(6):598–601. doi: 10.1158/2326-6066.CIR-15-0035. [DOI] [PubMed] [Google Scholar]
- 70*.Singer S, Grommes C, Reiner AS, Rosenblum MK, DeAngelis LM. Posterior reversible encephalopathy syndrome in patients with cancer. The Oncologist. 2015;20:806–811. doi: 10.1634/theoncologist.2014-0149. This study is the first large retrospective review of PRES in cancer patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71*.Kamiya-Matsouka C, Paker A, Chi L, Youssef A, Tummala S, Loghin ME. Posterior reversible encephalopathy syndrome in cancer patients: a single institution retrospective study. J Neurooncol. 2016 doi: 10.1007/s11060-016-2078-0. [Epub ahead of print]. This is the largest and most recent study of PRES in cancer patients. [DOI] [PubMed] [Google Scholar]
- 72.Maur M, Tomasello C, Frassoldati A, Dieci MV, Barbierei E, Conte P. Posterior reversible encephalopathy syndrome during ipilimumab therapy for malignant melanoma. J Clin Oncol. 2012;30(6):76–78. doi: 10.1200/JCO.2011.38.7886. [DOI] [PubMed] [Google Scholar]
- 73.Crona A, Whang YE. Posterior reversible encephalopathy syndrome induced by enzalutamide in a patient with castration-resistant prostate cancer. Invest New Drugs. 2015;33(3):751–754. doi: 10.1007/s10637-014-0193-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Eryilmaz MK, Mutlu H, Salim DK, Musri FY, Coskun HS. Fatal posterior reversible leukoencephalopathy syndrome associated coma induced by bevacizumab in metastatic colorectal cancer and review of the literature. J Oncol Pharm Pract. 2015 doi: 10.1177/1078155215611048. [DOI] [PubMed] [Google Scholar]
- 75.LaPorte J, Solh M, Ouanounou S. Posterior reversible encephalopathy syndrome following pembrolizumab therapy for relapsed Hodgkin’s lymphoma. J Oncol Pharm Pract. 2015 doi: 10.1177/1078155215620922. [DOI] [PubMed] [Google Scholar]
- 76.Munoz J, Kumar VA, Hamilton J, et al. Posterior reversible encephalopathy syndrome: more than meets the eye. J Clin Oncol. 2013;31(20):e360–363. doi: 10.1200/JCO.2012.46.6573. [DOI] [PubMed] [Google Scholar]
- 77.Carson KR, Evens AM, Richey EA, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood. 2009;113(20):4834–4840. doi: 10.1182/blood-2008-10-186999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wathes R, Moule S, Milojkovic D. Progressive multifocal leukoencephalopathy associated with ruxolitinib. N Engl J Med. 2013;369(2):197–198. doi: 10.1056/NEJMc1302135. [DOI] [PubMed] [Google Scholar]
- 79.Carson KR, Newsome SD, Kim EJ, et al. Progressive multifocal leukoencephalopathy associated with brentuximab vedotin therapy: a report of 5 cases from the Southern Network on Adverse Reactions (SONAR) project. Cancer. 2014;120(16):2464–2471. doi: 10.1002/cncr.28712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Lee HC, Mulanovich V, Nieto Y. Progressive multifocal leukoencephalopathy after allogeneic bone marrow transplantation for acute myeloid leukemia. J Natl Compr Canc Netw. 2014;12(12):1660–1664. doi: 10.6004/jnccn.2014.0167. quiz 1664. [DOI] [PubMed] [Google Scholar]
- 81.Bossolasco S, Calori G, Moretti F, et al. Prognostic significance of JC virus DNA levels in cerebrospinal fluid of patients with HIV-associated progressive multifocal leukoencephalopathy. Clin Infect Dis. 2005;40(5):738–744. doi: 10.1086/427698. [DOI] [PubMed] [Google Scholar]