Skip to main content
The Indian Journal of Medical Research logoLink to The Indian Journal of Medical Research
. 2018 Sep;148(3):254–257. doi: 10.4103/ijmr.IJMR_615_18

Pancreatic ductal adenocarcinoma: Role of chemotherapy & future perspectives

Akash Tiwari 1, Lalit Kumar 1,*
PMCID: PMC6251265  PMID: 30425214

The management of pancreatic ductal adenocarcinoma (PDAC) is a therapeutic challenge. Locally advanced/metastatic disease at presentation and significant genetic heterogeneity, a potential reason for resistance to cytotoxic chemotherapy are major barriers1. Incidence (per 100,000) of PDAC is low in India, 1.4 in males and 1.0 among females, compared to North America where corresponding figures are 7.5 and 6.5, respectively2. This burden of PDAC is likely to increase further in India with increase in longevity, changing lifestyle, increase in smoking/tobacco consumption, obesity and lack of physical activity/exercise - the key risk factors for this disease3,4.

The current treatment approach for PDCA patients with advanced/metastatic disease is to use chemotherapy; the choice of the regimen is based on the patient's fitness [Eastern Cooperative Oncology Group (ECOG) performance status], organ function, comorbidities, individual preference and psychosocial issues. Leucovorin plus short-term infusional fluorouracil plus oxaliplatin and irinotecan (FOLFIRINOX) is the preferred regimen for the young and fit patients [ECOG performance status (ECOG PS) 0, 1] with serum bilirubin <1.5 mg/dl5. FOLFIRINOX was compared to single-agent gemcitabine in a randomized phase III trial, which included patients with metastatic pancreatic cancer with ECOG PS 0, 1 and serum bilirubin <1.5 mg/dl. The patients received single-agent gemcitabine (1000 mg/m2 weekly 7 of 8 wk and then for 3 of 4 wk) and FOLFIRINOX (oxaliplatin, 85 mg/m2; irinotecan, 180 mg/m2; leucovorin, 400 mg/m2 and fluorouracil, 400 mg/m2 given as a bolus followed by 2400 mg/m2 as a 46 h continuous infusion, every 2 wk). The use of FOLFIRINOX was associated with a better overall response rate (ORR) (32 vs. 9%), progression-free survival (PFS) (6.4 vs. 3.3 months) and overall survival (OS) (11.1 vs. 6.8 months). Patients in combination arm had higher Grade 3/4 toxicity including febrile neutropenia, thrombocytopenia, neuropathy and diarrhoea5. It should be pointed out that only minorities of patients with metastatic pancreatic cancer are candidates for FOLFIRINOX due to older age and poor general condition. Patients who are candidates for intensive regimen and have serum bilirubin <1.5 mg/m2 but not a candidate for FOLFIRINOX are treated with a combination of gemcitabine and nab (nanoparticle albumin-bound) paclitaxel. In a phase III randomized controlled trial, MPACT, the combination of nab-paclitaxel (125 mg/m2) and gemcitabine (1000 mg/m2) for 3 wk every 4 weekly was compared to single-agent gemcitabine (1000 mg/m2 weekly 7 of 8 wk and then for 3 of 4 wk). The combination regimen was associated with a higher ORR (23 vs. 7%), PFS (5.5 vs. 3.7 months) and OS (8.5 vs. 6.7 months) and higher toxicity6. This study included patients older than 75 yr and with ECOG PS 2 which are not represented in the studies evaluating FOLFIRINOX. It has been suggested that nab-paclitaxel may deplete the tumour stroma through the binding of albumin to fibroblasts containing secreted protein acidic and rich in cysteine7.

In this issue Ostwal et al8 report a retrospective study evaluating the response and treatment outcome of patients with metastatic/locally advanced pancreatic cancer treated with a combination of gemcitabine and non-cremophor paclitaxel [abraxane (Abraxis BioScience, Inc., Los Angeles, CA, USA) or nanoxel (Dabur Pharma Ltd., India)]. The conventional paclitaxel is insoluble in water and hence cremophor; a castor oil-based surfactant is used to make it soluble. Cremophor contributes to the hypersensitivity reactions associated with paclitaxel infusion. The non-cremophor paclitaxel, abraxane is albumin-bound nanoparticles of paclitaxel while nanoxel is a polymeric-micelle formulation. The use of non-cremophor paclitaxel compared to conventional paclitaxel has an added advantage of lesser hypersensitivity reaction and lesser need for pre-medications. The authors report an ORR of 30.8 per cent, median PFS of 5.6 months [95% confidence interval (CI) 3.7-7.4] and a median OS of 11.6 months (95% CI 8.8-14.3) months. The OS and PFS in this study are comparable to that in the MPACT trial6. There was no significant difference in the OS and PFS among patients treated with gemcitabine-abraxane and gemcitabine-nanoxel combination. There was no difference in the treatment outcomes of these two-drug combinations as the study was not designed or powered to answer this question. Although it is undoubted that availability of nanoxel gives a cheaper option for patients who are to be treated with non-cremophor paclitaxel, which may be an important factor in a resource-limited setting. The other interesting point was that the grade III/IV toxicity in this study was lower than that reported in another phase III study6 evaluating this combination, which as per authors could be due to the dose modification incorporated in the treatment protocol. This gives a useful insight into means of limiting toxicity without affecting the efficacy and treatment outcome by tailoring the dose of chemotherapy regimen for an individual patient in a palliative setting. Other means have also been used to reduce the toxicity of this regimen, for example, administering gemcitabine (1000 mg/m2) and nab-paclitaxel (125 mg/m2) every two weeks. This regimen has comparable efficacy and reduced toxicity as compared to standard dosing schedule of nab-paclitaxel-gemcitabine9. Patients who are fit for combination chemotherapy but who continue to have bilirubin >1.5 mg/dl despite biliary stenting can be treated with FOLFOX regimen10. Other combination regimens have also been evaluated, for example, gemcitabine-5-fluorouracil and gemcitabine-capecitabine which have a doubtful benefit over single-agent gemcitabine11,12,13,14.

In view of the dismal outcome of patients with advanced pancreatic cancer several novel approaches have been evaluated. Pancreatic cancer has been shown to express epidermal growth factor receptor15. The vascular endothelial growth factor (VEGF) is also deemed important in the pathogenesis and spread of pancreatic cancer16. In keeping with these findings, a phase III study compared the gemcitabine (1000 mg/m2 weekly) with or without erlotinib (100 mg daily)17. Patients in the combination arm (gemcitabine and erlotinib) had a significant though small OS benefit of two weeks (6.2 vs. 5.9 months). This clinically insignificant benefit comes at a greater financial cost and toxicity. The combination of gemcitabine with VEGF inhibitor, for example, bevacizumab, or multikinase inhibitors, for example, axitinib or sorafenib, on the other hand, failed to show any benefit over single-agent gemcitabine17,18.

The patients who are not candidate for intensive chemotherapy and ECOG PS ≤2 can be offered single-agent gemcitabine (1000 mg/m2 weekly 7 of 8 wk and then for 3 of 4 wk)19. The use of single-agent gemcitabine is associated with low ORR (11%) with clinical benefit observed in nearly 30 per cent of the patients19. Clinical benefit was defined by decrease in pain, analgesic use and weight gain. This discordance between response assessed by response evaluation criteria in solid tumors (RECIST) criteria (recist.eortc.org)19 and clinical benefit may be due to the extensive desmoplastic reaction seen in pancreatic cancer which makes assessment of response at primary site difficult. Immunotherapy is another option for patients with metastatic pancreatic cancer with high microsatellite instability20.

Palliative care forms the bedrock for an appropriate management of metastatic/locally advanced metastatic PDAC. Care must be taken for adequate analgesia, nutrition, management of obstructive jaundice, gastric outlet obstruction and psychosocial well-being of these patients. Patients with significant comorbidities and a poor performance status should be referred for palliative care.

Genetically, PDAC is a very heterogeneous disease1; important mutations are KRAS (>90%), p53 (60-70%), CDKN2A, (>50%), SMAD4 (approximately 50%). About 4-5 per cent of patients have mutations in BRCA1/2; these cases have enhanced sensitivity to platinum-based chemotherapy as well as poly (ADP-ribose) polymerase (PARP) inhibition1. This improved understanding has led to initiation of a number of clinical trials (https://www.cancer.gov/types/pancreatic). Several new drugs such as methyl ethyl ketone (MEK) inhibitors, pegvorhyaluronidase alpha (PEGH20) which degrades hyaluronic acid in tumour microenvironment (TME), napabucasin (BBI-608), which inhibits cancer stem cells, and AM0010, a PEGylated interleukin-10 are at various stages of drug development and are being evaluated in combination with various agents21,22,23,24. Similarly, poly(ADP-ribose) polymerase (PARP) inhibitors are being evaluated as single-agent or as maintenance therapy in breast cancer (BRCA)-mutated patients with metastatic pancreatic cancer who have had a stable disease or objective response to chemotherapy25.

In PDCA, the stroma is very dense, fibrotic and heterogeneous and is possibly responsible for resistance to therapy. Recent progress in the field of immunotherapy and checkpoint inhibitors have shown promise in a number of cancers particularly in melanoma, lung cancer, urinary bladder cancer, renal cancer and head and neck cancer. Results of single-agent checkpoint inhibitors have not met with success. Possibly lower levels of neoantigens, the unique immunosuppressive TME and low levels of intratumoral infiltrating T-lymphocytes are some of the barriers26. Currently, a number of phase I/II trials are underway to evaluate the use of checkpoint inhibitors along with gemcitabine or targeted therapies for modulating and enhancing T-cell immunity26. Results of these trials are awaited with hope.

Footnotes

Conflicts of Interest: None.

References

  • 1.Mueller S, Engleitner T, Maresch R, Zukowska M, Lange S, Kaltenbacher T, et al. Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature. 2018;554:62–8. doi: 10.1038/nature25459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86. doi: 10.1002/ijc.29210. [DOI] [PubMed] [Google Scholar]
  • 3.Lowenfels AB, Maisonneuve P. Epidemiology and risk factors for pancreatic cancer. Best Pract Res Clin Gastroenterol. 2006;20:197–209. doi: 10.1016/j.bpg.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • 4.Michaud DS, Giovannucci E, Willett WC, Colditz GA, Stampfer MJ, Fuchs CS, et al. Physical activity, obesity, height, and the risk of pancreatic cancer. JAMA. 2001;286:921–9. doi: 10.1001/jama.286.8.921. [DOI] [PubMed] [Google Scholar]
  • 5.Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–25. doi: 10.1056/NEJMoa1011923. [DOI] [PubMed] [Google Scholar]
  • 6.Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–703. doi: 10.1056/NEJMoa1304369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sahoo RK, Kumar L. Albumin-bound paclitaxel plus gemcitabine in pancreatic cancer. N Engl J Med. 2014;370:478–9. doi: 10.1056/NEJMc1314761. [DOI] [PubMed] [Google Scholar]
  • 8.Ostwal V, Sahu A, Zanwar S, Nayak L, Shrikhande SV, Shetty N, et al. Experience with non-cremaphor based paclitaxel-gemcitabine regimen in advanced pancreatic cancer: Results from a single tertiary cancer centre. Indian J Med Res. 2018;148:284–90. doi: 10.4103/ijmr.IJMR_249_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Krishna K, Blazer MA, Wei L, Ahn DH, Wu CSY, Ciombor KK, et al. Modified gemcitabine and nab-paclitaxel in patients with metastatic pancreatic cancer (MPC): A single-institution experience. J Clin Oncol. 2015;33(Suppl 3):366. [Google Scholar]
  • 10.Ghosn M, Farhat F, Kattan J, Younes F, Moukadem W, Nasr F, et al. FOLFOX-6 combination as the first-line treatment of locally advanced and/or metastatic pancreatic cancer. Am J Clin Oncol. 2007;30:15–20. doi: 10.1097/01.coc.0000235997.18657.a6. [DOI] [PubMed] [Google Scholar]
  • 11.Berlin JD, Catalano P, Thomas JP, Kugler JW, Haller DG, Benson AB, 3rd, et al. Phase III study of gemcitabine in combination with fluorouracil versus gemcitabine alone in patients with advanced pancreatic carcinoma: Eastern Cooperative Oncology Group Trial E2297. J Clin Oncol. 2002;20:3270–5. doi: 10.1200/JCO.2002.11.149. [DOI] [PubMed] [Google Scholar]
  • 12.Di Costanzo F, Carlini P, Doni L, Massidda B, Mattioli R, Iop A, et al. Gemcitabine with or without continuous infusion 5-FU in advanced pancreatic cancer: A randomised phase II trial of the Italian oncology group for clinical research (GOIRC) Br J Cancer. 2005;93:185–9. doi: 10.1038/sj.bjc.6602640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Herrmann R, Bodoky G, Ruhstaller T, Glimelius B, Bajetta E, Schüller J, et al. Gemcitabine plus capecitabine compared with gemcitabine alone in advanced pancreatic cancer: A randomized, multicenter, phase III trial of the Swiss group for clinical cancer research and the central European Cooperative Oncology Group. J Clin Oncol. 2007;25:2212–7. doi: 10.1200/JCO.2006.09.0886. [DOI] [PubMed] [Google Scholar]
  • 14.Cunningham D, Chau I, Stocken DD, Valle JW, Smith D, Steward W, et al. Phase III randomized comparison of gemcitabine versus gemcitabine plus capecitabine in patients with advanced pancreatic cancer. J Clin Oncol. 2009;27:5513–8. doi: 10.1200/JCO.2009.24.2446. [DOI] [PubMed] [Google Scholar]
  • 15.Fjällskog ML, Lejonklou MH, Oberg KE, Eriksson BK, Janson ET. Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res. 2003;9:1469–73. [PubMed] [Google Scholar]
  • 16.Itakura J, Ishiwata T, Shen B, Kornmann M, Korc M. Concomitant over-expression of vascular endothelial growth factor and its receptors in pancreatic cancer. Int J Cancer. 2000;85:27–34. doi: 10.1002/(sici)1097-0215(20000101)85:1<27::aid-ijc5>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
  • 17.Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: A phase III trial of the national cancer institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–6. doi: 10.1200/JCO.2006.07.9525. [DOI] [PubMed] [Google Scholar]
  • 18.Kindler HL, Niedzwiecki D, Hollis D, Sutherland S, Schrag D, Hurwitz H, et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: Phase III trial of the cancer and leukemia group B (CALGB 80303) J Clin Oncol. 2010;28:3617–22. doi: 10.1200/JCO.2010.28.1386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Rothenberg ML, Moore MJ, Cripps MC, Andersen JS, Portenoy RK, Burris HA, 3rd, et al. A phase II trial of gemcitabine in patients with 5-FU-refractory pancreas cancer. Ann Oncol. 1996;7:347–53. doi: 10.1093/oxfordjournals.annonc.a010600. [DOI] [PubMed] [Google Scholar]
  • 20.Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20. doi: 10.1056/NEJMoa1500596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hingorani SR, Zheng L, Bullock AJ, Seery TE, Harris WP, Sigal DS, et al. HALO 202: Randomized phase II study of PEGPH20 plus nab-paclitaxel/gemcitabine versus nab-paclitaxel/gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma. J Clin Oncol. 2018;36:359–66. doi: 10.1200/JCO.2017.74.9564. [DOI] [PubMed] [Google Scholar]
  • 22.Hubbard JM, Grothey A. Napabucasin: An update on the first-in-class cancer stemness inhibitor. Drugs. 2017;77:1091–103. doi: 10.1007/s40265-017-0759-4. [DOI] [PubMed] [Google Scholar]
  • 23.Naing A, Papadopoulos KP, Autio KA, Ott PA, Patel MR, Wong DJ, et al. Safety, antitumor activity, and immune activation of pegylated recombinant human interleukin-10 (AM0010) in patients with advanced solid tumors. J Clin Oncol. 2016;34:3562–9. doi: 10.1200/JCO.2016.68.1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Matera R, Saif MW. New therapeutic directions for advanced pancreatic cancer: Cell cycle inhibitors, stromal modifiers and conjugated therapies. Expert Opin Emerg Drugs. 2017;22:223–33. doi: 10.1080/14728214.2017.1362388. [DOI] [PubMed] [Google Scholar]
  • 25.Kamel D, Gray C, Walia JS, Kumar V. PARP inhibitor drugs in the treatment of breast, ovarian, prostate and pancreatic cancers: An update of clinical trials. Curr Drug Targets. 2018;19:21–37. doi: 10.2174/1389450118666170711151518. [DOI] [PubMed] [Google Scholar]
  • 26.Rosenberg A, Mahalingam D. Immunotherapy in pancreatic adenocarcinoma-overcoming barriers to response. J Gastrointest Oncol. 2018;9:143–59. doi: 10.21037/jgo.2018.01.13. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Indian Journal of Medical Research are provided here courtesy of Scientific Scholar

RESOURCES