Skip to main content
Therapeutic Advances in Medical Oncology logoLink to Therapeutic Advances in Medical Oncology
editorial
. 2020 Feb 22;12:1758835920905412. doi: 10.1177/1758835920905412

‘The same old story’: thoughts on authorized doses of anticancer drugs

Fausto Meriggi 1,, Alberto Zaniboni 2
PMCID: PMC7036487  PMID: 32127926

Introduction

Worldwide, tumours are currently one of the main causes of death with approximately 9.6 million deaths a year and an incidence of approximately 18.1 million new cases in 2018.1 Consequently, the prevention, diagnosis and treatment of cancer represent, more than ever before, goals towards which all international administrations are moving; however, the results are not always tangible and comforting. One of the causes of these total or partial failures has been identified as the absence of adequate nationwide awareness programmes regarding the lifestyles most likely to reduce the incidence of cancer, as well as a shortage of funding for research and for the distribution of the most innovative medicinal products to the whole population. On the other hand, cancer is a huge business opportunity for pharmaceutical companies around the world, even more now than in the past. A review by Hong and colleagues2 shows that in the United States between 2011 and 2016, spending on cancer drugs grew by over 50% (from US$26.8 to 42.1 billion). Although the introduction of immune checkpoint inhibitors has dramatically changed the treatment of a number of cancer types, they bring a host of new adverse effects to be managed and a further exponential increase in both direct and indirect costs. Researchers are exploring new strategies making it possible to deal in the years to come with what can be defined as an authentic politico-socio-economic emergency that is now well identified, also regarding the possibly over simplistic terminology such as the ‘financial toxicity’ of cancer treatments. The price, not merely in economic terms, that is likely to have to be paid is dramatic meaning that much of the world population, even in higher income countries, could be excluded from access to the most novel cancer treatments. One well consolidated, albeit far from decisive, pathway undertaken in an attempt to reduce costs is the authorization of biosimilars and generic medicines. Another option to be given careful attention is that of reconsidering (at equal efficacy) the authorized doses of certain medicinal products, especially those with higher costs, particularly when used on a large scale such as those indicated for the treatment of the most frequently terminal cancers.

A few examples of more or less recent oncological agents that have undergone changes to their authorized regimens or that, in clinical practice, are almost always administered using posologies that differ from those originally authorized are reported in the following.

Abiraterone Acetate (AA) administered at a once-daily dose of 1000 mg on an empty stomach is the current standard of care for the treatment of hormone sensitive metastatic prostate cancer.36 An interesting article published recently in the Journal of Clinical Oncology based on the hypothesis that administering a dose of AA equal to one-quarter (250 mg) of the standard dose with a light, low fat breakfast is equally efficacious, showed its noninferiority in terms of both prostate-specific antigen response and time to progression compared with the authorized dose. The incidence of side effects was also similar.7 This finding could pave the way for the exploration of other alternative regimens in which AA is administered with food, such as a dose of 500 mg every other day, or even once every 4 days. It would appear unnecessary to stress the significant pharmacoeconomic implications that could be derived from this result.

Niraparib (N) is a poly (adenosine diphosphate-ribose) polymerase inhibitor approved in the USA and Europe for maintenance treatment of adult patients with recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer who are in complete or partial response to platinum based chemotherapy. In the pivotal study, N was administered at a once-daily dose of 300 mg.8 However, approximately 70% of patients had to reduce this dose due to adverse events and about 15% had to discontinue treatment, mainly for grade >3 thrombocytopenia. The retrospective study conducted by Berek and colleagues showed that, after adjustments to the dose of N, 200 mg is the dose most often used. Their analysis concluded that in patients weighing <77 kg (presumably the majority) or with platelet values <150,000 mm3 at baseline, N administered at a dose of 200 mg is able to achieve the same results, but with a far more acceptable toxicity profile.9

Lapatinib (L) is another example of how food is able to significantly change the bioavailability of a medicinal product. L was approved by the Food and Drug Administration (FDA) in 2007 in combination with capecitabine for advanced HER2-positive breast cancer in progression after previous treatments including anthracyclines, taxanes and trastuzumab. The regimen envisaged administering L at a flat dose of 1250 mg a day an hour before or an hour after breakfast, continuously.10 However, when L is administered with food, the (geometric) mean increase for the area under the concentration–time curve was 167% for low fat meals and 325% for high fat meals. These results are not surprising, given that food often increases a drug’s bioavailability, thereby making it possible to administer a drug at lower doses.11 Authors have gone so far as to make the provocative suggestion that L could be administered with food at 1/5 of the standard dose (250 mg instead of 1250 mg) and could be taken with a glass of grapefruit juice.12

Regorafenib is currently indicated for the treatment of metastatic colorectal cancer after at least two lines of therapy,13 for gastrointestinal stromal tumours (GIST) following progression on treatment with imatinib and sunitinib, and for hepatocellular cancer following progression on treatment with sorafenib, at a dose of 160 mg a day for 3 weeks, followed by a 1-week break in 28-day cycles.14 Gastrointestinal (GI), skin and hepatic toxicity often make it necessary to reduce the dose or even discontinue the treatment in heavily pretreated patients. The reDOS trial showed that a dose escalation of 40 mg a week starting from a dose of 80 mg makes it possible to limit toxicity, whilst maintaining efficacy, with an improvement in overall survival in the investigational arm.15

Ceritinib (C) 750 mg fasted is approved for treatment of patients with ALK receptor tyrosine kinase gene (ALK)-rearranged (ALK-positive) non-small cell lung cancer (NSCLC) previously treated with crizotinib.16 In an attempt to reduce the entity of gastrointestinal toxicity whilst maintaining the same pharmacokinetic and efficacy profile, part one of the ASCEND-8 study determined whether administering C 450 mg or 600 mg with a low fat meal can enhance GI tolerability versus 750 mg fasted in patients with ALK-positive NSCLC while maintaining similar exposure.17 This study demonstrated that C 450 mg administered with food has a similar exposure to the medicinal product and a GI toxicity profile that is considerably more favourable than C 750 mg in fasted patients with ALK-positive NSCLC. On the other hand, with the 600 mg dose, again administered with food, the steady state pharmacokinetics showed 25% higher levels, and it would therefore not appear to be suited to the aim of limiting toxicity.

Sunitinib is currently indicated for GISTs that are refractory to imatinib.18 and metastatic renal cell carcinoma (MRCC) with a recommended starting dose of 50 mg to be administered orally once daily, for four consecutive weeks, followed by a 2-week break (4/2 regimen) in cycles with an overall duration of 6 weeks.19 It is also indicated for the treatment of pancreatic neuroendocrine tumours; however, in this case, the recommended dose is a once-daily oral administration of 37.5 mg, without break periods.20 However, in GISTs and MRCC, following the considerable toxicities reported that usually force approximately 50% of patients to reduce the starting dose, in recent years, clinicians have increasingly opted for an alternative regimen at the same daily dose, but administered for 2 weeks followed by a week’s break (2/1 regimen). In the study by Bracarda and colleagues in MRCC, the shift due to toxicity from the 4/2 regimen to the 2/1 regimen led to a reduction in >grade 3 toxicity from 45.7% to 8.2%. More specifically, fatigue, hypertension, hand-foot syndrome and thrombocytopenia were less frequent.21 However, recently a panel of experts ruled that in MRCC, due to the methodological limits of the studies considered and, therefore, in the absence of certain efficacy data for the 2/1 regimen, despite the better tolerability observed it is still preferable to start therapy with the standard 4/2 regimen.22

Vinorelbine (V) is administered in the treatment of advanced breast cancer after failure of standard therapy, as a single agent or in combination, and as a first-line treatment for advanced NSCLC, as a single agent or in combination. When used as both a single agent and in combination, V is currently usually administered at a dose of 25–30 mg/m2 on days 1 and 8 of 21-day cycles; however, in the original authorized regimen, which dates back some 25 years, this agent was administered at the once-weekly dose of 30 mg/m2 continuously until disease progression or unacceptable toxicity.23,24

Capecitabine is currently indicated as adjuvant therapy for stage III bowel cancer, for the treatment of metastatic colorectal cancer, as a first-line therapy for advanced gastric cancer in combination with a regimen containing platinum, in combination with docetaxel for the treatment of patients with locally advanced or metastatic breast cancer following the failure of a chemotherapy regimen containing an anthracycline, as monotherapy for the treatment of patients with locally advanced or metastatic breast cancer following the failure of a chemotherapy regimen containing taxanes and an anthracycline, or for which further anthracycline therapy is not indicated. When used as a single agent, the recommended starting dose is 2500 mg/m2/day in two oral administrations on a full stomach (breakfast and dinner) for 14 days followed by a 1-week break, in 21-day cycles. In combination therapy, the recommended daily dose is 1600–2000 mg/m2/day, or 1250 mg/m2/day when administered continuously, as in the case of concomitant radiotherapy or metronomic chemotherapy. Two doses of capecitabine are currently available on the market: the 150 mg tablets (very rarely used) and the 500 mg tablets, which are practically the only kind used in clinical practice. This now consolidated habit by clinicians of using the 500 mg formulation alone, has led to approximations (often rounding down) of the overall amount of capecitabine that should be administered.25,26

Bevacizumab is an antivascular endothelial growth factor monoclonal antibody indicated in combination with a number of chemotherapy agents for a broad spectrum of advanced tumours (ovarian, breast, lung, cervical, renal and colorectal cancers). For example, in the treatment of colorectal cancer, in which this medicinal product is extensively used, the recommended dose of bevacizumab, is 5 mg/kg or 10 mg/kg once every other week, or 7.5 mg/kg or 15 mg/kg of body weight once every 3 weeks, depending on the regimen used.2730 The doses most frequently used in clinical practice are 5 mg/kg every other week and 7.5 mg/kg every 3 weeks.

Cabazitaxel (Cab) in combination with prednisone or prednisolone is indicated for the treatment of patients with castration-resistant metastatic prostate cancer who have previously been treated with a regimen containing docetaxel. The recommended dose is 25 mg/m2 administered every 3 weeks.31 However, one recently published phase III study showed the noninferiority of Cab at a 20% lower dose with an approximately 15% lower incidence of serious adverse events (primarily medullary toxicity, febrile neutropenia and diarrhoea).32

Docetaxel (D) is approved as a single agent (100 mg/m2 q21),33,34 or in combination in the adjuvant treatment of breast cancer,35 in advanced breast cancer3638 and as a second-line therapy for advanced NSCLC (75 mg/m2 q21)39,40 in combination with prednisone. It is also approved in the treatment of patients with metastatic prostate cancer (75 mg/m2 q21)41 in combination with cisplatin and 5-fluorouracil for the treatment of patients with metastatic cancer of the stomach and gastro–oesophageal junction who have not received prior treatment for the metastatic disease42 and, finally, again in combination with cisplatin and 5-fluorouracil, for the induction treatment of patients with locally advanced squamous cell carcinoma of the head and neck.4345 Nevertheless, in clinical practice, in order to minimize toxicity, docetaxel is now also often administered at a dose of 60 mg/m2 q21,46 or with the weekly regimen of 30–40 mg/m2/w for six consecutive weeks every 8 weeks47,48 or even with a biweekly regimen of 50 mg/m2.49

Liposomal doxorubicin is approved for the treatment of ovarian cancer, breast cancer, multiple myeloma, and Kaposi sarcoma.5052 In monochemotherapy it is still authorized at a dose of 50 mg/m2 every 28 days; however, in clinical practice the dose that is usually administered is 20% lower (40 mg/m2), with a significant reduction in the incidence and severity of palmar-plantar erythrodysesthesia and mucosites.53

Nivolumab, an anti-PD-1 monoclonal antibody, represents the most recent example of how an authorized dose (3 mg/kg every other week) can be altered without being based on a solid scientific rationale, but for primarily financial reasons. Nivolumab is indicated for the treatment of a number of solid tumours such as melanoma, renal cell carcinoma, NSCLC, urothelial cancer, squamous cell carcinoma of the head and neck, hepatocellular carcinoma, colorectal carcinoma and in haematological settings in Hodgkin’s lymphoma.5462 The strange thing is that since its marketing authorization, there has been a switch from using a dose of 3 mg/kg to a flat dose every 2 or 4 weeks (Table 1) thanks to debatable studies able to demonstrate an equivalence of efficacy.63,64 A recent editorial by Ratain and Goldstein65 states that there is significant pharmacokinetic and pharmacodynamic evidence supporting the theory that nivolumab can work just as well, and probably with a better immune-correlated toxicity profile, even at considerably lower doses. In a phase I study in 2012, nivolumab at 0.1 mg /kg (about 3% of 3 mg/kg) every other week showed activity and the ability to saturate receptors.66,67 This means that there is most likely no dose–response correlation and that nivolumab could probably be administered at doses considerably lower than those currently used without prejudicing the results.

Table 1.

Authorized doses of nivolumab administered as monotherapy (nivolumab technical files).

Indication Authorized doses Initial authorized dose Phase I proposed dose
Melanoma 240 mg q14 or 480 mg q28 3 mg/Kg q14 0.1 mg/kg q14
RCC 240 mg q14 or 480 mg q28 3 mg/Kg q14 0.1 mg/kg q14
NSCLC 240 mg q14 3 mg/Kg q14 0.1 mg/kg q14
Classic HD 240 mg q14 3 mg/Kg q14 0.1 mg/kg q14
SCC of H&N 240 mg q14 3 mg/Kg q14 0.1 mg/kg q14
Urothelial carcinoma 240 mg q14 3 mg/Kg q14 0.1 mg/kg q14

H&N, Head & Neck; HD, Hodgkin Disease; NSCLC, nonsmall cell lung cancer; RCC, renal cell carcinoma; SCC, Squamous Cell Carcinoma.

The same thing would appear to apply for pembrolizumab, another anti-PD-1 monoclonal antibody.68 For example, pembrolizumab monotherapy is currently administered at a flat dose of 200 mg every 3 weeks as first-line therapy for advanced NSCLC, in classic Hodgkin’s lymphoma and in urothelial cancer6971 and at 2 mg/kg every 3 weeks for NSCLC previously treated with chemotherapy and for melanoma.7274

Discussion

The history of medical oncology and haematology over the past 30 years has taught us that countless medicinal products, both chemotherapy agents and biologicals, are now used in clinical practice at different, usually lower, doses or with less intensive regimens than those for which they were authorized and marketed (Table 2). This happens for many reasons, some known, others more obscure, and lies primarily in the frequent biases present in the pivotal studies that often enrol patients who are not representative of the real population that is subsequently treated with that given medicinal product or that given regimen. It is no coincidence that an increasing number of postmarketing ‘real world’ observational studies are being conducted. For example, the enrolment in these studies of the elderly population >70 years of age is almost always lacking.75 Otherwise the differences in the metabolism of the medicinal product between the different ethnic groups and different sexes are not considered76 and the conclusions they reach regarding the toxicity observed at the maximum tolerated dose (MTD) are often reported in an ambiguous manner using unclear terminology; for example, ‘Most patients had an acceptable adverse-event profile’, or ‘. . .has a manageable and mostly reversible safety profile’, or ‘. . .the tolerability was good overall’ and ‘Incomplete reporting that downplayed drug-related adverse events was identified in 43% of reports of cancer drug trials’,77 thereby generating the illusion of having identified the dose that is transversally best for all types of patient. Furthermore, a recent analysis showed that out of 101 pivotal studies on medicinal products approved by the FDA, a high incidence has been reported for even considerable changes once the study is under way to the sample size, the inclusion criteria and the primary endpoint. More specifically, 56 studies (equal to 55.4%) underwent a change in the sample size, 34 studies (equal to 33.7%) underwent changes to the inclusion criteria and 27 studies (equal to 26.7%) underwent a change in the primary endpoint. In the final publication, these changes were described in 39 (69.6%) out of the 56 cases with changes in the sample size, in 19 (55.9%) of the 34 cases with changes in the inclusion criteria, and in 10 out of the 27 studies (37.0%) in which the primary endpoint was modified.78 ‘Many drugs are now approved on the basis of whether they shrink the tumour or delay the time until the tumour grows, but they don’t necessarily help patients to live longer or better lives’. Consequently, it is also essential to assess the impact these medicinal products have on the quality of life. Therefore, the simple question we should be asking is: ‘Does the dose or regimen with which the medical product is marketed actually represent what is best for the patient in terms of efficacy and tolerability?’ The ‘old’ concept of MTD in phase I studies has most likely now been superseded and perhaps we should work on a new emerging concept, that of ‘minimum efficacious dose’, which is far closer to the real world setting. Indeed, in regards to targeted therapies, for example, the best way to determine the optimum dose is still unclear, although various alternative strategies to MTD have been explored. Most oncological agents have a strong dose–response correlation; minimal changes in the dose administered can lead to severe toxicity that can be life threatening in some patients, and to under dosing in others, such as to prejudice the result. Consequently, choosing the right dose is an aspect of great importance, especially in individuals with a potentially curable disease or in the adjuvant therapy setting. However, choosing the best dose is complicated by the fact that individuals have very varied abilities to metabolize and eliminate medicinal products. The most relevant pharmacokinetic parameter for exposure to a medicinal product is the area under the curve (AUC) of the plasma concentration × unit of time after the administration of a single dose. However, the AUC is influenced by both external factors, such as the dose of the drug or the regimen used, and by factors that are intrinsic to the patient, such as age, sex, weight, height and genetic factors such as the capacity to metabolize a drug or its clearance, which correlates closely with hepatic and renal function.79 Therefore, in the attempt to minimize the potential subjective variables, the dose of most chemotherapy agents is still calculated on the basis of body surface area (BSA). The situation is completely different for targeted therapies, a very diverse and confusing field in which some medicinal products can be administered on the basis of body weight alone (e.g. monoclonal antibodies such as cetuximab, bevacizumab, intravenous trastuzumab, ipililumab, ramucirumab, panitumumab), others at a flat dose, such as a number of oral tyrosine kinase-inhibitors (e.g. dabrafenib, vemurafenib, trametinib, gefitinib, erlotinib, afatinib) or the m-TOR inhibitors, others still are administered both at doses calculated on the basis of body weight and flat doses such as certain immunotherapy agents (pembrolizumab, nivolumab, atezolizumab) or some monoclonal antibodies (e.g. alemtuzumab, ofatumumab, pertuzumab, subcutaneous trastuzumab). Finally, others, like carboplatin, are administered with doses calculated on the basis of the AUC.80,81 One paradigmatic example is olaparib, a PARP inhibitor, whose biological activity, considered as the inhibition of the enzyme in the tumour tissue, has been used to identify the most appropriate dose. In phase I studies, the biological activity of olaparib was demonstrated at doses achieved with two daily administrations of over 100 mg and with an MTD of 600 mg.82,83 The final dose subsequently approved by the FDA was 400 mg twice daily. Finally, despite the lack of robust data in favour of the routine use of BSA for dose calculation and an increasing number of scientific publications that question the validity of this method for a number of conventional cytotoxic agents,26,8492 the dose of most chemotherapy drugs is still calculated on the basis of the BSA.9396 Unfortunately, for most cancer drugs dose calculation using BSA has a limited ability to consider the individual variability in the clearance of the agent after the administration of a single dose.97,98 Oncologists are now aware that the administration of a drug calculated using BSA alone can have a subjective variability of over 30%,81 whereas the dose calculated on the basis of clearance values can oscillate within a range of between 4- and 10-fold.99,100

Table 2.

Differences between authorized dose/schedule and ‘real world’ dose/schedule.

Drug Authorized dose/schedule (Ref.) ‘real world’ administered/suggested dose/schedule (Ref.)
Lapatinib 1250 mg/day po on an empty stomach10 250 mg/day po with food11,12
Regorafenib 160 mg/day po × 21 days q2813,14 80→120→160 mg/day po (increase of 40 mg weekly based on tolerance)15
Ceritinib 750 mg/day po on an empty stomach16 450–600 mg/day with food17
Sunitinib 50 mg/day po (‘4/2’ schedule)18,19 or 37.5 mg/day po, continuously20 50 mg/day po (‘2/1’ schedule)21,22
Niraparib 300 mg/day po, continuously8 200 mg/day po, continuously9
Abiraterone Acetate 1000 mg/day po on an empty stomach, continuously36 250 mg/day po with a light, low fat breakfast, continuously7
Vinorelbine 30 mg/mq/weekly iv, continuously 25–30 mg/mq iv day 1.8 q21
Capecitabine 2500 mg/mq/day po × 14 days q21 1600–2000 mg/mq/day po × 14 days q21 or 1250 mg/mq/day, continuously25,26
Cabazitaxel 25 mg/mq q2131 20 mg/mq q2132
Docetaxel 100 mg/mq q213345 60–75 mg/mq q21 or 35–40 mg/mq/wk × 6 weeks every 8 weeks4649
Liposomal Doxorubicin 50 mg/mq q285052 40 mg/mq q2853
Bevacizumab 5–10 mg/kg q14 or 7.5–15 mg/kg q212730 5 mg/kg q14 or 7.5 mg/kg q21
Nivolumab 240 mg q14 or 480 mg q285464 0.1 mg/kg q146567
Pembrolizumab 200 mg q21 or 2 mg/kg q216871 2 mg/kg q217274

po, orally; iv, intravenous.

In conclusion, the evidence available shows that we all too often see a posteriori changes in the doses of authorized regimens of a number of cancer drugs without there being any negative repercussions on costs; rather, there is often a gain in terms of tolerability and in cost savings. We are therefore of the opinion that we need to rethink phase I dose-finding studies based on the identification of the MTD and in the reporting of toxicity in general and serious adverse events in particular, by introducing the ‘minimum efficacious dose’ concept.

Footnotes

Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.

Conflict of interest statement: The authors declare that there is no conflict of interest.

ORCID iD: Fausto Meriggi Inline graphic https://orcid.org/0000-0003-0292-340X

Contributor Information

Fausto Meriggi, Oncology Department, Poliambulanza Foundation, Via Leonida Bissolati 57, Brescia, 25124, Italy.

Alberto Zaniboni, Oncology Department, Poliambulanza Foundation, Brescia, Italy.

References

  • 1. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68: 394–424. [DOI] [PubMed] [Google Scholar]
  • 2. Hong SJ, Li EC, Matusiak LM, et al. Spending on antineoplastic agents in the United States, 2011 to 2016. J Onc Practice 2018; 14: e683–e690. [DOI] [PubMed] [Google Scholar]
  • 3. de Bono JS, Logothetis CJ, Molina A, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med 2011; 364: 1995–2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Ryan CJ, Smith MR, de Bono JS, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med 2013; 368: 138–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Fizazi K, Tran N, Fein L, et al. Abiraterone plus prednisone in metastatic, castration-sensitive prostate cancer. N Engl J Med 2017; 377: 352–360. [DOI] [PubMed] [Google Scholar]
  • 6. James ND, de Bono JS, Spears MR, et al. Abiraterone for prostate cancer not previously treated with hormone therapy. N Engl J Med 2017; 377: 338–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Szmulewitz RZ, Peer CJ, Ibraheem A, et al. Prospective international randomized phase II Study of low-dose abiraterone with food versus standard dose abiraterone in castration-resistant prostate cancer. J Clin Oncol 2018; 36: 1389–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med 2016; 375: 2154–2164. [DOI] [PubMed] [Google Scholar]
  • 9. Berek JS, Matulonis UA, Peen U, et al. Safety and dose modification for patients receiving Niraparib. Ann Oncol 2018; 29; 1784–1792. [DOI] [PubMed] [Google Scholar]
  • 10. Geyer CE, Forster J, Lindquist D, et al. Lapatinib plus Capecitabine for HER2-positive advanced breast cancer. N Engl J Med 2006; 355: 2733–2743. [DOI] [PubMed] [Google Scholar]
  • 11. Devriese LA, Koch KM, Mergui-Roelvink M, et al. Effects of low-fat and high-fat meals on steady-state pharmacokinetics of lapatinib in patients with advanced solid tumours. Invest New Drugs 2014; 32: 481–488. [DOI] [PubMed] [Google Scholar]
  • 12. Ratain MJ, Cohen EE. The value meal: how to save $1700 per month or more on Lapatinib. J Clin Oncol 2007; 25: 3397–3398. [DOI] [PubMed] [Google Scholar]
  • 13. Grothey A, Van Cutsem E, Sobrero A, et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicenter, randomized, placebo-controlled, phase 3 trial. Lancet 2013; 381: 303–312. [DOI] [PubMed] [Google Scholar]
  • 14. Bruix J, Qin S, Merle P, et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017; 389: 56–66. [DOI] [PubMed] [Google Scholar]
  • 15. Bekaii-Saab TS, Ou FS, Anderson DM, et al. Regorafenib dose optimization study (ReDos). A phase II randomized study of lower dose regorafenib compared to standard dose regorafenib in patients with refractory metastatic colorectal cancer (mCRC). J Clin Oncol 2018; 36(Suppl. 4S): Abstract 611. [Google Scholar]
  • 16. Shaw AT, Kim TM, Crinò L, et al. Ceritinib versus chemotherapy in patients with ALK-rearranged non-small-cell lung cancer previously given chemotherapy and crizotinib (ASCEND-5): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol 2017; 18: 874–886. [DOI] [PubMed] [Google Scholar]
  • 17. Cho BC, Kim DW, Bearz A, et al. ASCEND-8: A randomized phase I study of Ceritinib, 450 mg or 600 mg, taken with a low-fat meal versus 750 mg in fasted state in patients with anaplastic lymphoma kinase (ALK)-rearranged metastatic non-small cell lung cancer (NSCLC). J Thorac Oncol 2017; 12: 1357–1367. [DOI] [PubMed] [Google Scholar]
  • 18. Demetri GD, van Oosterom AT, Garrett CR, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet 2006; 368: 1329–1338. [DOI] [PubMed] [Google Scholar]
  • 19. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007; 356: 115–124. [DOI] [PubMed] [Google Scholar]
  • 20. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 2011; 364: 501–513. [DOI] [PubMed] [Google Scholar]
  • 21. Bracarda S, Iacovelli R, Boni L, et al. Sunitinib administered on 2/1 schedule in patients with metastatic renal cell carcinoma: the RAINBOW analysis. Ann Oncol 2015; 26: 2107–2113. [DOI] [PubMed] [Google Scholar]
  • 22. Bracarda S, Negrier S, Casper J, et al. How clinical practice is changing the rules: the sunitinib 2/1 schedule in metastatic renal cell carcinoma. Expert rev Anticancer Ther 2017; 17: 227–233. [DOI] [PubMed] [Google Scholar]
  • 23. Depierre A, Lemarie E, Dabouis G, et al. A phase II study of Navelbine (vinorelbine) in the treatment of non-small-cell lung cancer. Am J Clin Oncol 1991; 14: 115–119. [DOI] [PubMed] [Google Scholar]
  • 24. Le Chevalier T, Brisgand D, Douillard JY, et al. Randomized study of vinorelbine and cisplatin versus vindesine and cisplatin versus vinorelbine alone in advanced non-small-cell lung cancer: results of a European multicenter trial including 612 patients. J Clin Oncol 1994; 12: 360–367. [DOI] [PubMed] [Google Scholar]
  • 25. Ratain MJ. Editorial: “Dear Doctor: we really are not sure what dose of capecitabine you should prescribe for your patient”. J Clin Oncol 2002; 20: 1434–1435. [DOI] [PubMed] [Google Scholar]
  • 26. Cassidy J, Twelves C, Cameron D, et al. Bioequivalence of two tablet formulations of capecitabine and exploration of age, gender, body surface area, and creatinine clearance as factors influencing systemic exposure in cancer patients. Cancer Chemother Pharmacol 1999; 44: 453–460. [DOI] [PubMed] [Google Scholar]
  • 27. Hochster HS, Hart LL, Ramanathan RK, et al. Safety and efficacy of oxaliplatin and fluoropyrimidine regimens with or without bevacizumab as first- line treatment of metastatic colorectal cancer: results of the TREE study. J Clin Oncol 2008; 26: 3523–3529. [DOI] [PubMed] [Google Scholar]
  • 28. Saltz LB, Clarke S, Dıaz-Rubio E, et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol 2008; 26: 2013–2019. [DOI] [PubMed] [Google Scholar]
  • 29. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004; 350: 2335–2342. [DOI] [PubMed] [Google Scholar]
  • 30. Goldstein DA, Chen Q, Ayer T, et al. First- and second-line bevacizumab in addition to chemotherapy for metastatic colorectal cancer: a United States-based cost-effectiveness analysis. J Clin Oncol 2015; 33: 1112–1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. de Bono JS, Oudard S, Ozguroglu M, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet 2010; 376: 1147–1154. [DOI] [PubMed] [Google Scholar]
  • 32. Eisenberger M, Hardy-Bessard AC, et al. Phase III study comparing a reduced dose of cabazitaxel (20 mg/mq) and the currently approved dose (25 mg/mq) in post-docetaxel patients with metastatic castration-resistant prostate cancer - PROSELICA. J Clin Oncol 2017; 35: 3198–3206. [DOI] [PubMed] [Google Scholar]
  • 33. Francis P, Crown J, Di Leo A, et al. Adjuvant chemotherapy with sequential or concurrent anthracycline and docetaxel: breast international group 02–98 randomized trial. J Natl Cancer Inst 2008; 100: 121–133. Eiermann W, Pienkowski T, Crown J, et al. Phase III study of doxorubicin/cyclophosphamide with concomitant versus sequential docetaxel as adjuvant treatment in patients with human epidermal growth factor receptor 2- normal, node-positive breast cancer: BCIRG-005 trial. J Clin Oncol 2011; 29: 3877–3884. [Google Scholar]
  • 34. Jones SE, Savin MA, Holmes FA, et al. Phase III trial comparing doxorubicin plus cyclophosphamide with docetaxel plus cyclophosphamide as adjuvant therapy for operable breast cancer. J Clin Oncol 2006; 24: 5381–5387. [DOI] [PubMed] [Google Scholar]
  • 35. Dear RF, McGeechan K, Jenkins MC, et al. Combination versus sequential single agent chemotherapy for metastatic breast cancer. Cochrane Database Syst Rev 2013; 12: CD00879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. O’Shaughnessy J, Miles D, Vukelja S, et al. Superior survival with capecitabine plus docetaxel combination therapy in anthracycline-pretreated patients with advanced breast cancer: phase III trial results. J Clin Oncol 2002; 20: 2812–2823. [DOI] [PubMed] [Google Scholar]
  • 37. Jones S, Holmes FA, O’Shaughnessy J, et al. Docetaxel with cyclophosphamide is associated with an overall survival benefit compared with doxorubicin and cyclophosphamide: 7-year follow-up of US oncology research trial 9735. J Clin Oncol 2009; 27: 1177–1183. [DOI] [PubMed] [Google Scholar]
  • 38. Zalcberg J, Millward M, Bishop J, et al. Phase II study of docetaxel and cisplatin in advanced non-small-cell lung cancer. J Clin Oncol 1998; 16: 1948–1953. [DOI] [PubMed] [Google Scholar]
  • 39. Shepherd FA, Dancey J, Ramlau R, et al. Prospective randomized trial of docetaxel versus best supportive care in patients with non-small-cell lung cancer previously treated with platinum-based chemotherapy. J Clin Oncol 2000; 18: 2085–2103. [DOI] [PubMed] [Google Scholar]
  • 40. Fossella FV, DeVore R, Kerr RN, et al. Randomized phase III trial of docetaxel versus vinorelbine or ifosfamide in patients with non-small cell lung cancer previously treated with platinum - containing chemotherapy regimens. J Clin Oncol 2000; 18: 2354–2362. [DOI] [PubMed] [Google Scholar]
  • 41. Tannock IF, de Wit R, Berry WR, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 2004; 351: 1502–1512. [DOI] [PubMed] [Google Scholar]
  • 42. Van Cutsem E, Moiseyenko VM, Tjulandin S, et al. Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group. Lancet Oncol 2006; 24: 4991–4997. [DOI] [PubMed] [Google Scholar]
  • 43. Posner MR, Hershock DM, Blajman CR, et al. Cisplatin and fluorouracil alone or with docetaxel in head and neck cancer. N Engl J Med 2007; 357: 1705–1715. [DOI] [PubMed] [Google Scholar]
  • 44. Vermorken JB, Remenar E, van Herpen C, et al. Cisplatin, fluorouracil, and docetaxel in unresectable head and neck cancer. N Engl J Med 2007; 357: 1695–1704. [DOI] [PubMed] [Google Scholar]
  • 45. Paccagnella A, Ghi MG, Loreggian L, et al. Concomitant chemoradiotherapy versus induction docetaxel, cisplatin and 5 fluorouracil (TPF) followed by concomitant chemoradiotherapy in locally advanced head and neck cancer: a phase II randomized study. Ann Oncol 2010; 21: 1515–1522. [DOI] [PubMed] [Google Scholar]
  • 46. Nakamura Y, Kunitoh H, Kubota K, et al. Retrospective analysis of safety and efficacy of low-dose docetaxel 60 mg/mq in advanced non-small cell lung cancer patients previously treated with platinum-based chemotherapy. Am J Clin Oncol 2003; 26: 459–464. [DOI] [PubMed] [Google Scholar]
  • 47. Stemmler J, Mair W, Stauch M, et al. High efficacy and low toxicity of weekly docetaxel given as first-line treatment for metastatic breast cancer. Oncology 2005; 68: 71–78. [DOI] [PubMed] [Google Scholar]
  • 48. Tabernero J, Climent MA, Lluch A, et al. A multicentre, randomised phase II study of weekly or 3-weekly docetaxel in patients with metastatic breast cancer. Ann Oncol 2004; 15: 1358–1365. [DOI] [PubMed] [Google Scholar]
  • 49. Kellokumpu-Lehtinen PL, Harmenberg U, Joensuu T, et al. 2-Weekly versus 3-weekly docetaxel to treat castration-resistant advanced prostate cancer: a randomised, phase 3 trial. Lancet Oncol 2013; 14: 117–124. [DOI] [PubMed] [Google Scholar]
  • 50. Markman M, Kennedy A, Webster K, et al. Phase 2 trial of liposomal doxorubicin (40 mg/mq) in platinum/paclitaxel-refractory ovarian and fallopian tube cancers and primary carcinoma of the peritoneum. Gynecol Oncol 2000; 78: 369–372. [DOI] [PubMed] [Google Scholar]
  • 51. Perez AT, Domenech GH, Frankel C, et al. Pegylated liposomal doxorubicin (Doxil) for metastatic breast cancer: the Cancer Research Network, Inc., experience. Cancer Invest 2002; 20 (Suppl. 2): 22–29. [DOI] [PubMed] [Google Scholar]
  • 52. O’Brien ME, Wigler N, Inbar M, et al. Reduced cardiotoxicity and comparable efficacy in a phase III trial of pegylated liposomal doxorubicin HCl (CAELYX/Doxil) versus conventional doxorubicin for first-line treatment of metastatic breast cancer. Ann Oncol 2004; 15: 440–449. [DOI] [PubMed] [Google Scholar]
  • 53. Al-Batran SE, Meerpohl HG, von Minckwitz G, et al. Reduced incidence of severe palmar-plantar erythrodysesthesia and mucositis in a prospective multicenter phase II trial with pegylated liposomal doxorubicin at 40 mg/mq every 4 weeks in previously treated patients with metastatic breast cancer. Oncology 2006; 70: 141–146. [DOI] [PubMed] [Google Scholar]
  • 54. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined nivolumab and ipililumab or monotherapy in untreated melanoma. N Engl J Med 2015; 373; 23–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Weber J, Mandala M, Del Vecchio M, et al. Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma. N Engl J Med 2017; 377: 824–835. [DOI] [PubMed] [Google Scholar]
  • 56. Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with metastatic DNA mismatch repair deficient/microsatellite instability–high colorectal cancer (CheckMate 142): results of an open-label, multicentre, phase 2 study. Lancet Oncol 2017; 18: 1182–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. El-Khoueiry AB, Sangro B, Yau T, et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017; 389: 2492–2502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non–small-cell lung cancer. N Engl J Med 2015; 373: 123–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non–small-cell lung cancer. N Engl J Med 2015; 373: 1627–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med 2015; 373: 1803–1813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Ferris RL, Blumenschein G, Fayette J, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med 2016; 375: 1856–1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Herbaux C, Gauthier J, Brice P, et al. Efficacy and tolerability of nivolumab after allogeneic transplantation for relapsed Hodgkin lymphoma. Blood 2017; 129: 2471–2478. [DOI] [PubMed] [Google Scholar]
  • 63. Zhao X, Ivaturi V, Gopalakrishnan M, et al. A model-based exposure-response (E-R) assessment of a nivolumab (NIVO) 4-weekly (every 4 weeks) dosing schedule across multiple tumor types. Presented at American Association for Cancer Research Annual Meeting 2017, 1–5 April 2017, Washington, DC. [Google Scholar]
  • 64. Zhao X, Suryawanshi S, Hruska M, et al. Assessment of nivolumab benefit- risk profile of a 240-mg flat dose relative to a 3-mg/kg dosing regimen in patients with advanced tumors. Ann Oncol 2017; 28: 2002–2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Ratain MJ, Goldstein DA. Time is money: optimizing the scheduling of nivolumab. J Clin Oncol 2018; 36: 3074–3076. [DOI] [PubMed] [Google Scholar]
  • 66. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366: 2443–2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Topalian SL, Sznol M, McDermott DF, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 2014; 32: 1020–1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Patnaik A, Kang SP, Rasco D, et al. Phase I study of pembrolizumab (MK-3475; anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. Clin Cancer Res 2015; 21: 4286–4293. [DOI] [PubMed] [Google Scholar]
  • 69. Reck M, Rodriguez-Abreu D, Robinson AG, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non–small-cell lung cancer. N Engl J Med 2016; 375: 1823–1833. [DOI] [PubMed] [Google Scholar]
  • 70. Chen R, Zinzani PL, Fanale MA, et al. Phase II study of the efficacy and safety of pembrolizumab for relapsed/refractory classic Hodgkin lymphoma. J Clin Oncol 2017; 35: 2125–2132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Bellmunt J, Vaughn DJ, Fradet Y, et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017; 376: 1015–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1- positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016; 387: 1540–1550. [DOI] [PubMed] [Google Scholar]
  • 73. Robert C, Schachter J, Long GV, et al. Pembrolizumab versus Ipilimumab in advanced melanoma. N Engl J Med 2015; 372: 2521–2532. [DOI] [PubMed] [Google Scholar]
  • 74. Ribas A, Puzanov I, Dummer R, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol 2015; 16: 908–918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Arciero VS, Cheng S, Mason R, et al. Do older and younger patients derive similar survival benefits from novel oncology drugs. A systematic review and meta-analysis. Age Ageing 2018; 47: 654–660. [DOI] [PubMed] [Google Scholar]
  • 76. Davidson M. Influence of sex on chemotherapy efficacy and toxicity in oesophagogastric (OG) cancer: a pooled analysis of 4 randomised trials. Presented at ESMO 2018 Congress, 19 October 2018, Munich, Germany. Abstract 619PD_PR. [DOI] [PubMed] [Google Scholar]
  • 77. Gyawali B, Shimokata T, Honda K, et al. Reporting harms more transparently in trials of cancer drugs. Br Med J 2018; 363: k4383. [DOI] [PubMed] [Google Scholar]
  • 78. Shepshelovich D, Tibau A, Molto C, et al. Assessment of frequency and reporting of changes in cancer trial design after initiation of patient accrual. JAMA Oncol. Epub ahead of print 6 December 2018. DOI: 10.1001/jamaoncol.2018.5877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Thummel KE, Lin YS. Sources of interindividual variability. Methods Mol Biol 2014; 1113: 363–415. [DOI] [PubMed] [Google Scholar]
  • 80. Schellens JH, Beijnen JH. Novel clinical trial designs for innovative therapies. Clin Pharmacol Ther 2009; 85: 212–216. [DOI] [PubMed] [Google Scholar]
  • 81. Sparreboom A, Verweij J. Advances in cancer therapeutics. Clin Pharmacol Ther 2009; 85: 113–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Fong PC, Boss DS, Yap TA, et al. Inhibition of poly (ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 2009; 361: 123–134. [DOI] [PubMed] [Google Scholar]
  • 83. Fong PC, Yap TA, Boss DS, et al. Poly (ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J Clin Oncol 2010; 28: 2512–2519. [DOI] [PubMed] [Google Scholar]
  • 84. Grochow LB, Baraldi C, Noe D. Is dose normalization to weight or body surface area useful in adults? J Natl Cancer Inst 1990; 82: 323–325. [DOI] [PubMed] [Google Scholar]
  • 85. de Jongh FE, Verweij J, Loos WJ, et al. Body-surface area-based dosing does not increase accuracy of predicting cisplatin exposure. J Clin Oncol 2001; 19: 3733–3739. [DOI] [PubMed] [Google Scholar]
  • 86. Dobbs NA, Twelves CJ. What is the effect of adjusting epirubicin doses for body surface area? Br J Cancer 1998; 78: 662–666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Etienne MC, Chatelut E, Pivot X, et al. Co-variables influencing 5-fluorouracil clearance during continuous venous infusion. A NONMEM analysis. Eur J Cancer 1998; 34: 92–97. [DOI] [PubMed] [Google Scholar]
  • 88. Mathijssen RH, Verweij J, de Jonge MJ, et al. Impact of body-size measures on irinotecan clearance: alternative dosing recommendations. J Clin Oncol 2002; 20: 81–87. [DOI] [PubMed] [Google Scholar]
  • 89. Teresi ME, Riggs CE, Webster PM, et al. Bioequivalence of two methotrexate formulations in psoriatic and cancer patients. Ann Pharmacother 1993; 27: 1434–1438. [DOI] [PubMed] [Google Scholar]
  • 90. Loos WJ, Gelderblom H, Sparreboom A, et al. Inter- and intrapatient variability in oral topotecan pharmacokinetics: implications for body-surface area dosage regimens. Clin Cancer Res 2000; 6: 2685–2689. [PubMed] [Google Scholar]
  • 91. Freyer G, Tranchand B, Ligneau B, et al. Population pharmacokinetics of doxorubicin, etoposide and ifosfamide in small cell lung cancer patients: results of a multicentre study. Br J Clin Pharmacol 2000; 50: 315–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Ratain MJ, Mick R, Schilsky RL, et al. Pharmacologically based dosing of etoposide: a means of safely increasing dose intensity. J Clin Oncol 1991; 9: 1480–1486. [DOI] [PubMed] [Google Scholar]
  • 93. Baker SD, Verweij J, Rowinsky EK, et al. Role of body surface area in dosing of investigational anticancer agents in adults, 1991–2001. J Natl Cancer Inst 2002; 94: 1883–1888. [DOI] [PubMed] [Google Scholar]
  • 94. Sawyer M, Ratain MJ. Body surface area as a determinant of pharmacokinetics and drug dosing. Invest New Drugs 2001; 19: 171–177. [DOI] [PubMed] [Google Scholar]
  • 95. Ratain MJ. Body-surface area as a basis for dosing of anticancer agents: science, myth, or habit? J Clin Oncol 1998; 16: 2297–2298. [DOI] [PubMed] [Google Scholar]
  • 96. Smorenburg CH, Sparreboom A, Bontenbal M, et al. Randomized cross-over evaluation of body-surface area-based dosing versus flat-fixed dosing of paclitaxel. J Clin Oncol 2003; 21: 197–202. [DOI] [PubMed] [Google Scholar]
  • 97. Felici A, Verweij J, Sparreboom A. Dosing strategies for anticancer drugs: the good, the bad and body-surface area. Eur J Cancer 2002; 38: 1677–1684. [DOI] [PubMed] [Google Scholar]
  • 98. Chatelut E, White-Koning ML, Mathijssen RH, et al. Dose banding as an alternative to body surface area-based dosing of chemotherapeutic agents. Br J Cancer 2012; 107: 1100–1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Gurney H. Dose calculation of anticancer drugs: a review of the current practice and introduction of an alternative. J Clin Oncol 1996; 14: 2590–2611 [DOI] [PubMed] [Google Scholar]
  • 100. Eaton KD, Lyman GH. Dosing of anticancer agents in adults. UpToDate, https://www.uptodate.com/contents/dosing-of-anticancer-agents-in-adults (accessed on 25 December, 2018).

Articles from Therapeutic Advances in Medical Oncology are provided here courtesy of SAGE Publications

RESOURCES