Metronomic temozolomide as second line treatment for metastatic poorly differentiated pancreatic neuroendocrine carcinoma

C De Divitiis, C von Arx, A M Grimaldi, D Cicala, F Tatangelo, A Arcella, G M Romano, E Simeone, R V Iaffaioli, P A Ascierto, S Tafuto, European Neuroendocrine Tumor Society (ENETS) Center of Excellence-Multidisciplinary Group for Neuroendocrine Tumors in Naples (Italy), C De Divitiis, C von Arx, A M Grimaldi, D Cicala, F Tatangelo, A Arcella, G M Romano, E Simeone, R V Iaffaioli, P A Ascierto, S Tafuto, European Neuroendocrine Tumor Society (ENETS) Center of Excellence-Multidisciplinary Group for Neuroendocrine Tumors in Naples (Italy)

Abstract

Neuroendocrine Neoplasms (NEN) are a group of heterogeneous malignancies derived from neuroendocrine cell compartment, with different roles in both endocrine and nervous system. Most NETs have gastroentero-pancreatic (GEP) origin, arising in the foregut, midgut, or hindgut. The 2010 WHO classification divides GEP-NETs into two main subgroups, neuroendocrine tumors (NET) and neuroendocrine carcinomas (NEC), according with Ki-67 levels. NET are tumors with low (<20 %) Ki-67 value, and NECs, including small cell lung carcinomas and Merkel Cell carcinomas, are all NETs with high Ki-67 levels (>20 %-G3). Poorly differentiated neuroendocrine carcinomas (NEC) are usually treated with cisplatin-based chemotherapy regimens. Here we present a case of a patient with pancreatic NEC progressing after cisplatin and etoposide, treated with temozolomide as palliative, second line treatment. According with the poor Performance Status (PS = 2) and to reduce the toxicity of the treatment was chosen an intermittent dosing regimen of metronomic temozolomide (75 mg/m(2)/day-one-week-on/on-week-off). MGMT resulted methylated. On July 2014 the patient started the treatment. On August 2014 the patient obtained a significant clinical benefit (PS = 0) and the total body CT scan performed on October 2014 showed a RECIST partial response on all the sites of disease. No drug-related side effects were reported by the patient. After 18 months of therapy the treatment continues without significant toxicity, and with further remission of the metastases. Treatment with metronomic "one-week-on/on-week-off" Temozolomide can be considered a good treatment option in patients with poor performance status, affected by pNEC with MGMT methylation.

Keywords: Immunotherapy; Metronomic treatment; Neuroendocrine tumors; Pancreatic neuroendocrine carcinoma; Second line therapy; Temozolomide.

Figures

Fig. 1
Fig. 1
Histological examination with immunohistochemistry and hematoxylin-eosin staining (e/e). a CD 56 20 × magnification; b e/e 20 × magnification; c chromogranin 20 × magnification; d Synaptophysin 20 × magnification; e Ki-67 40 × magnification
Fig. 2
Fig. 2
Peritumoral lymphocytes and leucocytes infiltrating tumor microenviroment. Tumor-infiltrating CD8 + T cells (circles) have been detected in the pNEC tumor biopsy of the patient. These cells infiltrating tumor micro environment are frequently associated with favorable clinical outcome in a remarkably large spectrum of cancers as MCC and pNEC
Fig. 3
Fig. 3
Detection of MGMT methylation by MSP and MethyLight qMSP. Isolated DNA from tumor specimen is treated with sodium bisulfite, which changes unmethylated cytosine (C) into uracil (U), but not methylated cytosine (mC), which remains unchanged. The modified DNA is used as a template for MSP or MethyLight qMSP. (MSP/agarose gel electrophoresis) When MGMT in neuroendocrine tumor is methylated (DNA M), the band is high, while the unmethylated band (DNA U) is low. The gel shows the presence of methylation band in BT1, brain tumor a with strong methylated, DNA M, used as positive control, and unmethylation band in BT2, brain tumor a with strong unmethylated, DNA U, used as negative control
Fig. 4
Fig. 4
Hepatomegaly reduction. CT portal-phase contrast-enhanced images: Coronal plane reconstruction and Axial Maximum Intensity Projection (MIP) reconstruction at time 0 (a) and at 4 (b), 7 (c), 10 month (d) follow up examinations. Progressive reduction of longitudinal diameter of the liver measured in the mid-clavicular line was observed at follow up study. Progressive reduction of lower margin extension below costal arch was also detected. Caudal displacement of right kidney at time 0 (white arrow) gradually disappears at successive studies. Axial MIP images show progressive reduction of the displacement effect on the portal bifurcation due to decrease of metastatic parenchymal lesions: space between left and right portal trunk (angle) gradually decrease over time. Note the increased caliber of portal trunks and progressive reduction of the swelling of liver profile (arrowhead) related with hepatomegaly, with increased thickness of perivisceral fat
Fig. 5
Fig. 5
CT portal-phase contrast-enhanced images of two different planes at time 0 (a) and at 4 (b), 7 (c), 10 month (d) follow up examinations. Conspicuous volume reduction of paraesophageal and paracardial lymph nodes was observed over time. Increased caliber of portal trunks was detected, due to parenchymal architecture changes, reduced mass effect and mild portal hypertension. Note progressive dislocation of left portal trunk asterisk from left to right side due to reduction of hepatomegaly. Also note volume decrease of liver and spleen lesions with gradually remission of parenchymal architecture
Fig. 6
Fig. 6
Changes of lesions over time in pancreatic head region, lung, liver and spleen at time 0 (a) and at 4 (b), 7 (c), 10 month (d) follow up examinations. Pancreas: enlargement of pancreatic head region was observed at time 0, with strongly inhomogeneous density, indistinct pancreatic margins and surrounding retroperitoneal fat stranding; some peripancreatic lymph nodes enlarged were detected. Following CT examinations show gradually decrease of pancreatic swelling with better definition of parenchymal lesions, going towards progressive regression e colliquation. Note progressive appearance of right kidney’s upper pole asterisk, according to reduction of displacement effect by liver. Furthermore, note the displacement of superior mesenteric artery (black arrow) to right side due to reduction of pancreatic swelling. Lung: progressive volume reduction of metastatic lesions (white arrow) in anterior basal segment of right lower pulmonary lobe. Liver: one of the major lesions of the liver at the IV segment (arrowhead) underwent progressive volume reducing ad enhancement pattern. Spleen: volume reduction of multiple metastatic lesions

References

    1. Williams ED, Sandler M. The classification of carcinoid tum ours. Lancet. 1963;1:238–239. doi: 10.1016/S0140-6736(63)90951-6.
    1. Yao JC, Hassan M, Phan A, et al. One hundred years after“carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26:3063–3072. doi: 10.1200/JCO.2007.15.4377.
    1. Gastrointestinal Pathology Study Group of Korean Society of Pathologists, Cho MY, Kim JM, et al Current trends of the incidence and pathological diagnosis of gastroenteropancreatic neuroendocrine tumors (GEPNETs)in Korea 2000–2009: multicenter study. Cancer Res Treat. 2012;44:157–165. doi: 10.4143/crt.2012.44.3.157.
    1. Bernick PE, Klimstra DS, Shia J, et al. Neuroendocrine carcinomas of the colon and rectum. Dis Colon Rectum. 2004;47:163–169. doi: 10.1007/s10350-003-0038-1.
    1. Milan SA, Yeo CJ. Neuroendocrine tumors of the pancreas. Curr Opin Oncol. 2012;24(1):46–55. doi: 10.1097/CCO.0b013e32834c554d.
    1. Yao JC, Eisner MP, Leary C, et al. Population-based study of islet cell carcinoma. Ann Surg Oncol. 2007;14:3492–3500. doi: 10.1245/s10434-007-9566-6.
    1. Rinke A, Müller H-H, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID study group. JCO. 2009;27(28):4656–4663. doi: 10.1200/JCO.2009.22.8510.
    1. Caplin ME, Pavel M, Ćwikła JB, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224–233. doi: 10.1056/NEJMoa1316158.
    1. Toumpanakis, et al. Cytotoxic treatment including embolization/chemoembolization for neuroendocrine tumours. Best Pract Res Clin Endocrinol Metab. 2007;21(1):131–144. doi: 10.1016/j.beem.2007.01.005.
    1. Nisa, et al. Yttrium-90 DOTATOC therapy in GEP-NET and other SST2 expressing tumors: a selected review. Ann Nucl Med. 2011;25(2):75–85. doi: 10.1007/s12149-010-0444-0.
    1. Moertel CG, Kvols LK, O’Connell MJ, et al. Treatment of neuro-endocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Abstr US Endocr Soc. 1991;68:227–232.
    1. Ridolfi L, Petrini M, Granato AM, et al. Low-dose temozolomide before dendritic-cell vaccination reduces (specifically) CD4+CD25++Foxp3+ regulatory T-cells in advanced melanoma patients. J Transl Med. 2013;11:135. doi: 10.1186/1479-5876-11-135.
    1. Kulke MH, Stuart K, Enzinger PC, Ryan DP, Clark JW, Muzikansky A, et al. Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. J Clin Oncol. 2006;24:401–406. doi: 10.1200/JCO.2005.03.6046.
    1. Ekeblad S, Sundin A, Janson ET, Welin S, Granberg D, Kindmark H, et al. Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clin Cancer Res. 2007;15:2986–2991. doi: 10.1158/1078-0432.CCR-06-2053.
    1. Kulke M, Blaszkowsky LS, Zhu AX, et al. Phase I/II study of everolimus (RAD001) in combination with temozolomide (TMZ) in patients (pts) with advanced pancreatic neuroendocrine tumors (NET). 2010 ASCO Gastrointestinal Cancers Symposium, January 22-24, 2010 (Abstract).
    1. Koumarianou A, Antoniou S, Kanakis G, Economopoulos N, Rontogianni D, Ntavatzikos A, et al. Combination treatment with metronomic temozolomide, bevacizumab and long-acting octreotide for malignant neuroendocrine tumours. Endocr Relat Cancer. 2012;19:L1–L4. doi: 10.1530/ERC-11-0287.
    1. Strosberg JR, Fine RL, Choi J, Nasir A, Coppola D, Chen DT, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Abstr US Endocr Soc. 2011;117:268–275.
    1. Welin S, Sorbye H, Sebjornsen S, Knappskog S, Busch C, Oberg K. Clinical effect of temozolomide-based chemotherapy in poorly differentiated endocrine carcinoma after progression on first-line chemotherapy. Abstr US Endocr Soc. 2011
    1. Gounaris I, Rahamim J, Shivasankar S, Earl S, Lyons B, Yiannakis D. Marked response to a cisplatin/docetaxel/temozolomide combination in a heavily pretreated patient with metastatic large cell neuroendocrine lung carcinoma. Anticancer Drugs. 2007;18:1227–1230. doi: 10.1097/CAD.0b013e32827bc61b.
    1. Bravo EL, Kalmadi SR, Gill I. Clinical utility of temozolomide in the treatment of malignant paraganglioma: a preliminary report. Horm Metab Res. 2009;41:703–706. doi: 10.1055/s-0029-1224135.
    1. Lindholm DP, Eriksson B, Granberg D. Response to temozolomide and bevacizumab in a patient with poorly differentiated neuroendocrine carcinoma. Med Oncol. 2012;29:301–303. doi: 10.1007/s12032-010-9789-4.
    1. Isacof WH, Moss RA, Pecora AL, Fine L. Temozolomide/capcitabine therapy for metastatic neuroendocrine tumors of the pancreas: a retrospective review. J Clin Oncol. 2006;24(20 Suppl):Abstr 14023.
    1. Strosberg JR, Fine RL, Choi J, Nasir A, Coppola D, Chen DT, Helm J, Kvols L. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117(2):268–275. doi: 10.1002/cncr.25425.
    1. Ott PA, Maria Elez-Fernandez ME, Hiret S, Kim DW, Moss RA, Winser T, et al. Pembrolizumab (MK-3475) in patients (pts) with extensive-stage small cell lung cancer (SCLC): Preliminary safety and efficacy results from KEYNOTE-028. J Clin Oncol. 2015;33:abstr 7502.
    1. Welin S, Sorbye H, Sebjornsen S, Knappskog S, Busch C, Oberg K. Clinical effect of temozolomide-based chemotherapy in poorly differentiated endocrine carcinoma after progression on first-line chemotherapy. Abstr US Endocr Soc. 2011
    1. Newlands ES, Blackledge GR, Slack JA, et al. Phase I trial of temozolomide(CCRG 81045: M&B 39831: NSC 362856) Br J Cancer. 1992;65:287–291. doi: 10.1038/bjc.1992.57.
    1. Newlands ES, Stevens MFG, Wedge SR, Wheelhouse RT, Brock C. Temozolomide: a review of its discovery, chemical properties, preclinical development and clinical trials. Cancer Treat Rev. 1997;23:35–61. doi: 10.1016/S0305-7372(97)90019-0.
    1. Wolfgang Wick, Michael Platten, and Michael Weller, New (alternative) temozolomide regimens for the treatment of glioma, Neuro-Oncology, 2009.
    1. Caroli M, Locatelli M, Campanella R, et al. Temozolomide in glioblastoma: results of administration at first relapse and in newly diagnosed cases. Is still proposable an alternative schedule to concomitant protocol? J Neurooncol. 2007;84:71–77. doi: 10.1007/s11060-007-9343-1.
    1. Chinot OL, Barrie M, Fuentes S, et al. Correlation between O6-methylguanine- DNA methyltransferase and survival in inoperable newly diagnosed glioblastoma patients treated with neoadjuvant temozolomide. J Clin Oncol. 2007;25:1470–1475. doi: 10.1200/JCO.2006.07.4807.
    1. Combs SE, Gutwein S, Schulz-Ertner D, et al. Temozolomide combined with irradiation as postoperative treatment of primary glioblastoma multiforme. Phase I/II study. Strahlenther Onkol. 2005;181:372–377. doi: 10.1007/s00066-005-1359-x.
    1. Combs SE, Schulz-Ertner D, Welzel T, Bischof M, Debus J. Re-irradiation using high precision radiotherapy and concomitant temozolomide in patients with recurrent glioma: re-challenge with radio-chemotherapy [abstract 12517] J Clin Oncol. 2007;25(suppl):606s.
    1. Tai-Gyu Kim,Chang-Hyun Kim,Jung-Sun Park,Sung-Dong Park,Chung Kwon Kim, Dong-Sup Chung,and Yong-Kil Hong Immunological Factors Relating to the Antitumor Effect of Temozolomide Chemoimmunotherapy in a Murine Glioma Model. Clin Vaccine Immunol. 2010;17:143–153. doi: 10.1128/CVI.00292-09.
    1. Nistico P, Capone I, Palermo B, Del Bello D, Ferraresi V, Moschella F, et al. Chemotherapy enhances vaccine-induced antitumor immunity in melanoma patients. Int J Cancer. 2009;124:130–139. doi: 10.1002/ijc.23886.
    1. Palermo B, Del Bello D, Sottini A, Serana F, Ghidini C, Gualtieri N, et al. Dacarbazine treatment before peptide vaccination enlarges T-cell repertoire diversity of melan-a- specific, tumor-reactive CTL in melanoma patients. Cancer Res. 2010;70:7084–7092. doi: 10.1158/0008-5472.CAN-10-1326.
    1. Poschke I, Mougiakakos D, Kiessling R. Camouflage and sabotage: tumor escape from the immune system. Cancer Immunol Immunother. 2011;60:1161–1171. doi: 10.1007/s00262-011-1012-8.
    1. Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T-cell-dependent antitumor immunity. Cancer Res. 2010;70:3052–3061. doi: 10.1158/0008-5472.CAN-09-3690.
    1. Mundy-Bosse BL, Lesinski GB, Jaime-Ramirez AC, Benninger K, Khan M, Kuppusamy P, et al. Myeloid-derived suppressor cell inhibition of the IFN response in tumor-bearing mice. Cancer Res. 2011;71:5101–5110. doi: 10.1158/0008-5472.CAN-10-2670.
    1. Banissi C, Ghiringhelli F, Chen L, Carpentier AF. Treg depletion with a low-dose metronomic temozolomide regimen in a rat glioma model. Cancer Immunol Immunother. 2009;58:1627–1634. doi: 10.1007/s00262-009-0671-1.
    1. Schiavoni G, Sistigu A, Valentini M, Mattei F, Sestili P, Spadaro F, et al. Cyclophosphamide synergizes with type I interferons through systemic dendritic cell reactivation and induction of immunogenic tumor apoptosis. Cancer Res. 2011;71:768–778. doi: 10.1158/0008-5472.CAN-10-2788.
    1. Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11:215–233. doi: 10.1038/nrd3626.

Source: PubMed

3
Subskrybuj