Open Access

Meta‑analysis of current chemotherapy regimens in advanced pancreatic cancer to prolong survival and reduce treatment‑associated toxicities

  • Authors:
    • Jie Chen
    • Linli Chen
    • Jianping Yu
    • Yanmei Xu
    • Xiaohui Wang
    • Ziqian Zeng
    • Ning Liu
    • Fan Xu
    • Shu Yang
  • View Affiliations

  • Published online on: November 9, 2018     https://doi.org/10.3892/mmr.2018.9638
  • Pages: 477-489
  • Copyright : © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Unresectable advanced pancreatic cancer (APC) is a highly lethal malignancy. Although numerous chemotherapeutic regimens are available, evidence regarding the survival extension, the life quality improvement, the associated risks and occurrence rates of adverse effects, is required. The effects of 19 chemotherapy regimens on survival and treatment‑associated toxicities in the context of APC treatment were comparatively assessed. A total of 23 randomized controlled trials were included in this network meta‑analysis. For overall survival, five regimens, Gemcitabine (Gem)+radiotherapy (Radio), Gem+cisplatin (Cis), Gem+erlotinib (Erl)+bevacizumab (Bev), Gem+capecitabine (Cap)+Erl, and Gem+exatecan, were the most effective treatments, according to their respective high surface under the cumulative ranking (SUCRA) probabilities. Regarding the progression‑free survival, five regimens, including Gem+Radio, Gem+Erl+Bev, Gem+Cis, Gem+Cap+Erl and Gem+pemetrexed, were the most effective treatments based on their SUCRA probabilities. Each regimen exhibited advantages and disadvantages, and 14 common treatment‑associated toxicities were present in different proportions. The three principal toxic effects included haematological, gastrointestinal and constitutional symptoms. To improve survival, chemotherapy regimens with high SUCRA probabilities require prioritizing. Although treatment‑associated toxicities are unavoidable, the regimens presented toxicities in distinct proportions. Therefore, clinicians should assess the disease status of the patients, and balance the benefits and risks of the selected treatment.

Introduction

Unresectable advanced pancreatic cancer (APC) is the most lethal and the most aggressive human cancer (1). APC is predicted to increase from the 4th to the 2nd leading cause of mortality in the USA by 2020 due to its lethal and malignant characteristics (2). Due to the limitations of diagnostic techniques, the majority of patients and clinicians become aware of the disease too late, as this cancer is frequently diagnosed in an advanced stage (3,4). APC is characterized by a high mortality rate worldwide, 90.8% in China (5), 78.5% in the USA (6) and 95.0% in Canada (7).

Gemcitabine (Gem) was more effective compared with 5-fluorouracil (5-FU) in patients with APC and improved the survival rate; therefore, it was approved as a first-line regimen by the US Food and Drug Administration (FDA) in 1996 (8). At present, the majority of chemotherapy regimens are derived from Gem, which was used as the control treatment in numerous previous studies (911).

Although a number of combination chemotherapy regimens containing Gem (Gem+Xs) or monotherapies have become more prevalent over the past decades (12), the improvement of the conditions of the patients has been limited (1215). For example, the poor prognosis of APC leads to a low survival rate (14,16,17), which has remained relatively unaltered for ~5 decades (15). Nevertheless, the benefits and risks of combination chemotherapy regimens remain unclear. Therefore, first-line chemotherapy regimen data were pooled to comprehensively evaluate the benefits and risks of these treatments.

Materials and methods

Study design

In order to assess the benefits and risks of various chemotherapy regimens in distinct conditions, head-to-head comparison clinical trials were selected. This network meta-analysis followed the preferred reporting items of system reviews and meta-analysis (PRISMA) statement (18) while integrating evidence from direct and indirect treatment comparisons (19). The flow chart for study selection is presented in Fig. 1.

Search strategy

The comprehensive search strategy was conducted using the MEDLINE (www.pubmed.com), EMBASE (www.embase.com), Cochrane Central Register of Controlled Trails (www.cochranelibrary.com) and ClinicalTrials.gov (https://ClinicalTrials.gov) databases with the following search terms: (Advanced pancreatic cancer or pancreatic cancer) AND (advanced pancreatic cancer or chemotherapy regimens). The drug abbreviations and the combinations tested are listed in Table I.

Table I.

Abbreviation list of chemistry regimens.

Table I.

Abbreviation list of chemistry regimens.

AbbreviationChemistry regimens
GemGemcitabine
Gem+Axit Gemcitabine+axitinib
Gem+5-FU Gemcitabine+5-fluorouracil
Gem+Cap+Erl Gemcitabine+capecitabin+erlotinib
Gem+Cap Gemcitabine+capecitabine
Gem+Cet Gemcitabine+cetuximab
Gem+Cis Gemcitabine+cisplatin
Gem+Erl Gemcitabine+erlotinib
Gem+Erl+Bev Gemcitabine+erlotinib+bevacizumab
Gem+Eta Gemcitabine+etanercept
Gem+Exa Gemcitabine+exatecan
Gem+Iri Gemcitabine+irinotecan
Gem+Mar Gemcitabine+marismastat
Gem+Nab-p Gemcitabine+nab-paclitaxel
Gem+Pem Gemcitabine+pemetrexed
Gem+Radio Gemcitabine+radiotherapy
Gem+Sor Gemcitabine+sorafenib
Gem+Tip Gemcitabine+tipifarnib
Gem+Vis Gemcitabine+vismodegib
Oxa+Iri+Leu+Flu+Inf Oxaliplatin+irinotecan+leucovorin+fluorouracil+infusion
Study selection criteria and outcomes

Experienced investigators independently selected the studies and extracted the data, and any conflicts were resolved in discussion. The study selection criteria were based on the National Comprehensive Cancer Network 2017 criteria (20). The following inclusion criteria were applied: (i) Parallel-group randomized controlled trials (RCTs; phase II or III) with the Gem intervention set as the common comparison treatment and including ≥2 arms; (ii) a minimum 6-month follow-up period; (iii) patients ≥18 years old (i.e., adult patients); (iv) diagnosis of unresectable APC; (v) application of palliative treatments, including invasive radiation therapy, chemotherapy or chemoradiation therapy, targeted therapies or combination therapy with the respective placebo or control group; (vi) either fixed-dose or flexible-dose RCTs with dose titration; and (vii) the patient performance status reported as 0–2 scores in the Eastern cooperative oncology group or 70–80% in Karnofsky scales (21). Previous studies that (i) included patients undergoing radical resection, (ii) failed to report the number of patients, (iii) failed to report the primary efficacy outcome [progression-free survival (PFS)], or (iv) failed to report the data necessary to estimate the standard deviation of the primary efficacy outcome were excluded.

Data extraction

The following data were extracted from each included RCT: First author's name, published year, clinical phase, sample size of each arm, age, treatment, dosage, route, duration, overall survival (OS) in months, PFS in months, and 14 treatment-associated categories of side effects associated with quality of life (‘hepatotoxicity’, ‘haematological’, ‘mental/psychiatry’, ‘renal toxicity’, ‘gastrointestinal’, ‘neuropathy’, ‘electrolytes imbalance’, ‘pain’, ‘infection’, ‘skin’, ‘constitutional symptoms’, ‘cardiac/vascular’, ‘pulmonary’ and ‘other’). The details of the outcomes of the included studies are presented in Table II.

Table II.

General characteristics of the included studies.

Table II.

General characteristics of the included studies.

Author, yearPhaseNMedian age, years (range)RegimensDoseRouteDuration, median (range) or mean ± SDOverall survival, months mean (95% CI)Progression-free survival, months mean (95% CI)(Refs.)
Cunningham et al, 2009326662 (26–83)GemGem (1,000 mg/m2/week) weeklyIV 6.23.8(30)
326762 (37–82)Gem+CapGem (1,000 mg/m2/week) + Cap 1,660 mg/m2/dayIV and oral 7.15.3
Conroy et al, 2005N/A17161 (34–75)GemGem (1,000 mg/m2/week)IV10 weeks6.8 (5.5–7.6)3.3 (2.2–3.6)(32)
N/A17161 (25–76) Oxa+Iri+Leu+FluOxi (85 mg/m2) + Iri (180 mg/m2 + Leu (400 mg/m2) + Flu (400 mg/m2)IV10 weeks11.1 (9.0–13.1)6.4 (5.5–7.2)
Berlin et al, 2002316264.3GemGem (1,000 mg/m2/week)IV3 weeks of every 45.42.2(41)
316065.8Gem+5-FUGem (1,000 mg/m2/week) + 5-FU (600 mg/m2/week)IV and bolus3 weeks of every 46.73.4
Bramhall et al, 2002N/A11962 (37–85)GemGem (1,000 mg/m2/week)IV10 weeks5.473.2(42)
N/A12062 (32–83)Gem+MarGem (1,000 mg/m2/week) + Mar 10 mg b.i.d.IV and bolus10 weeks5.523.08
Rocha Lima et al, 2004318060.2 (32.3–82.9)GemGem (1,000 mg/m2/week) (6.6–88)IV12.9 weeks6.6 (0.03–22.8)3.0 (2.5–3.7)(43)
318063.2 (38.7–81.2)Gem+IriGem (1,000 mg/m2/week) + Iri 100 mg/m2IV12.1 weeks (3.0–83.9)6.3 (0.2–23.8)3.5 (2.8–4.2)
Van Cutsem et al, 2004334762 (30–88)GemGem (1,000 mg/m2/week)IV14 weeks6.073.63(44)
334161 (29–89)Gem+TipGem (1,000 mg/m2/week) + Tip 200 mg b.i.d.IV and bolus12.1 weeks6.433.73
Oettle et al, 2005327363 (28–82)GemGem (1,250 mg/m2/week)IV12 weeks6.2 (5.4–6.9)3.3 (2.5–3.6)(45)
327363 (27–83)Gem+PemGem (1,000 mg/m2/week) + Pem 500 mg/m2IV12 weeks6.3 (5.4–6.9)3.9 (3.3–4.7)
Von Hoff et al, 2005343063GemGem (1,000 mg/m2/week)IV3 months6.73.7(46)
343163Gem+Nab-pN/AIV4 months8.55.5
Abou-Alfa et al, 2006317462.3 (30–84)GemGem (1,000 mg/m2/week)IV5.8±3.76.2 (5.2–7.5)3.8 (3–4.3)(47)
317563 (36–85)Gem+ExaGem (1,000 mg/m2/week) + Exa 2.0 mg/m2IVmonths 6.4±4.2 months6.7 (5.4–7.9)3.7 (2.7–4.7)
Heinemann et al, 200639766 (43–85)GemGem (1,000 mg/m2/week)IV4.1 months7.55.3(48)
39864 (37–82)Gem+CisGem (1,000 mg/m2/week) + Cis 50 mg/m2IV3.3 months63.1
Stathopoulos et al, 200637064 (44–83)GemGem (900 mg/m2/week)IV12 weeks6.52.9(49)
36064 (31–84)Gem+IrlGem (900 mg/m2/week) + Iri 300 mg/m2IV9 weeks6.42.8
Herrmann et al, 20073159N/AGemGem (1,000 mg/m2/week)IV 7.23.9(50)
3159N/AGem+CapGem (1,000 mg/m2/week) + Cap 650 mg/m2IV and oral 8.44.3
Moore et al, 2007328464.0 (36.1–92.4)GemGem (1,000 mg/m2/week)IV 5.913.55(51)
328563.7 (37.9–84.4)Gem+ErlGem (1,000 mg/m2/week) + Erl 100 mg/daysIV 6.243.75
Spano et al, 200823461 (36–78)GemGem 1,000 mg/m2 on days 1,8 and 15 in 4-week cyclesIV4 (1–12) cycles5.6 (3.9–8.8)3.7 (2.2–6.7)(52)
26965 (44–81)Gem+AxiGem 1,000 mg/m2 on day Axi 5 mg b.i.d.POAxitinib 113 (7–481) days,
Gemcitabine 5 (1–18) cycles6.9 (5.3–10.1)4.2 (3.6–10.2)
Colucci et al, 2010319963 (37–75)GemGem (1,000 mg/m2/week)IV8 cycles8.33.9(40)
320163 (35–75)Gem+CisGem (1,000 mg/m2/week) + Cis 25 mg/m2IV7 cycles7.23.8
Philip et al, 2010337164.3GemGem (1,000 mg/m2/week)IV 5.93(53)
337263.7Gem+CetGem (1,000 mg/m2/week) + Cet 400 mg/m2IV 6.33.4
Kindler et al, 2011331662 (35–89)GemGem (1,000 mg/m2/week) weeklyIV2.3 (0.03–11.0) months8.3 (6.9–10.3)4.4 (3.7–5.2)(54)
331461 (34–84)Gem+AxiGem (1,000 mg/m2/week) + Axi 10 mg/dayIV and oral2.8 (0.03–11.0) months8.5 (6.9–9.5)4.4 (4.0–5.6)
Loehrer et al, 2011N/A3767±8.7aGem1,000 mg/m2/week for weeks
69 (49.7–83.7) 1 to 6, followed by 1 week rest. Following rest, for 3 of 4 weeksIV5.5 (2–8.3) weeks9.2 (7.9–11.4)6.7(55)
N/A34 65.34±10.3a 66 (46.9–83.5)Gem+Radio600 mg/m2/weekIV5.5 (2–8.3) weeks11.1 (7.6–15.5)6
Heinemann et al, 2012N/A14365 (32–78)GemGem (1,000 mg/m2/week)IV5 cycles (0–26)6.93.2(56)
N/A14163 (38–75)Gem+Cap+ ErlGem (1,000 mg/m2/week) + Cap (1,000 mg/m2 twice daily) + Erl (150 mg daily)IV5 cycles (0–26)6.22.2
Wu et al, 20131/2859 (46–75)GemGem 1,000 mg/m2 weekly for 7 weeks with a one-week rest, followed 1,000 mg/m2 weekly for 3 weeks' with a one-week restIV12.8 (8–22) weeks8.1 (3.1–20.4)4.3 (2.2–8.1)(57)
1/23059 (46–81)Gem+EtaEta 25 mg twice weeklySubcutaneous12.2 (2–40) weeks5.43 (1.5–16.9)2.23 (1.8–7.4)
Moehler et al, 201424864.5 (36–84)GemGem (1,000 mg/m2) was administered on days 1, 8, 15, 22, 29, 36 and 43 of the first cycle (8 weeks duration) and days 1, 8 and 15 of all subsequent cycles (4 weeks duration)IV4.2 (0.3–21.4) months4.9 (3.5–7.7)4.9 (3.5–7.7)(58)
24964.0 (44–83)Gem+SorSor 400 mgIV and PO2.3 (0–19.1) months3 (1.8–7.2)3 (1.8–7.2)
Catenacci et al, 20151b/25364 (39–84)GemGem 1,000 mg/m2 IV over 30 min on days 1, 8 and 15 every 28 daysIV3 (0–14) cycles6.1 (5.0–8.0)2.5 (1.9–3.8)(59)
1b/25364 (49–82)Gem+VisGDC-0449 150 mg daily4 (1–12) cycles6.9 (5.8–8.0)4.0 (2.5–5.3)
Ramanathan et al, 2016343063 (32–88)GemGem 1,000 mg/m2 weekly for 7 of 8 weeks (cycle 1); in subsequent cycles, all patients were administered treatment on days 1, 8 and 15 every 4 weeksIV 6.7 (6.0–7.2)3.7 (3.6–4.0)(60)
343162 (27–86)Gem+Nab-pGem 1,000 mg/m2 on days 1, 8, 15, 29, 36 and 43 + Nab-P 1+++25 mg/m2IV 8.5 (7.9–9.5)5.5 (4.5–5.9)

a Mean ± SD. SD, standard deviation; N/A, not applicable; b.i.d., twice daily; IV, intravenous; PO, per os; CI, confidence interval; Gem, gemcitabine; Axit, axitinib; 5-FU, 5-fluorouracil; Erl, erlotinib; Cap, capecitabine; Cet, cetuximab; Cis, cisplatin; Bev, bevacizumab; Eta, etanercept; Exa, exatecan; Iri, irinotecan; Mar, marismastat; Nab-p, nab-paclitaxel; Pem, pemetrexed; Radio, radiotherapy; Sor, sorafenib; Tip, tipifarnib; Vis, vismodegib; Oxa, oxaliplatin; Leu, leucovorin; Flu, fluorouracil.

Risk of bias assessment

To reduce the risk bias, the recommended approach of Cochrane reviews was followed and the risk was assessed throughout the process (22). The following bias sources were independently assessed: Random sequence generation, allocation concealment, blinding of investigators and/or patients, blinding of outcome assessment and the degree of data incompleteness. Each bias was scored as low, unclear or high, as presented in Table III.

Table III.

Risk assessment of the included studies.

Table III.

Risk assessment of the included studies.

Author, yearRandom sequence generationAllocation concealmentBlinding of participants and personnelBlinding of assessmentIncomplete outcome outcome dataSelective reportingOtherbiasTotal scores(Refs.)
Cunningham et al, 2009LLLLHUL5(30)
Conroy et al, 2005UHLLLUL4(32)
Berlin et al, 2002LLLLLLL7(41)
Bramhall et al, 2002LLLLLLL7(42)
Rocha Lima et al, 2004LLHLLLL6(43)
Van Cutsem et al, 2004LLLLLUL6(44)
Oettle et al, 2005LLLLLLL7(45)
Von Hoff et al, 2005LLLLLLL7(46)
Abou-Alfa et al, 2006LLLLLLL7(47)
Heinemann et al, 2006LLLLLLL7(48)
Stathopoulos et al, 2006LLLLLLL7(49)
Herrmann et al, 2007LLLLHLL6(50)
Moore et al, 2007LLLLHLL6(51)
Spano et al, 2008LLLLLLL7(52)
Colucci et al, 2010LLLLLLL7(40)
Philip et al, 2010LLLLHLL7(53)
Kindler et al, 2011LLLLLLL7(54)
Loehrer et al, 2011LLLLLLL7(55)
Heinemann et al, 2012LLLLLLL7(56)
Wu et al, 2013LLLLLLL7(57)
Moehler et al, 2014LLLLLLL7(58)
Catenacci et al, 2015LLLLLLU6(59)
Ramanathan et al, 2016LLLLHLL6(60)

[i] L, low risk of bias; U, unclear risk of bias; H, high risk of bias.

Statistical analysis

All statistical analyses were performed using the network meta-analysis package in Stata (version 13.0; StataCorp LP, College Station, TX, USA) (19). For the endpoint outcomes, OS and PFS data were extracted from references as medians and subsequently transformed into standardized mean differences with 95% confidence intervals (CIs). A network meta-analysis was conducted following the standard workflow (19). The network map presents the connection status of the studies; no loops and a P-value >0.05 validated the consistency model to perform the network meta-analysis. Heterogeneity was assessed with the I2 metric. Heterogeneity was 0% for OS and 75.64% for PFS. Therefore, the Mantel-Haenszel fixed-effects model was used for OS, and the Mantel-Haenszel random-effects model was used for PFS (21). All of the studies with various treatments were included as drugs that were either directly or indirectly associated with a common comparator (Gem only) for further downstream ranking analysis. Subsequently, to rank the effects of the treatments, the analysis of the surface under the cumulative ranking (SUCRA) probabilities was performed under the protocol of Stata (19), and the results are presented as the percentage of the efficacy of each intervention relative to a hypothetical ideal intervention (23). A larger SUCRA score indicated longer OS and PFS.

The data are presented in an ordinal data format according to the 14 categories of side effects. Network meta-analyses were separately conducted, and the data were calculated as hazard ratios with 95% CIs. To examine and classify the adverse effects that occurred among the different treatments, a stack bar graph of each category was generated.

Results

Standard workflow via PRISMA

To ensure the general quality of the present study, a PRISMA flowchart regarding the screening process of the study used, is presented in Fig. 1. From an initial set of 7,855 non-duplicated studies, a total of 23 RCTs were included in this analysis. The drug abbreviations and the combinations assessed are listed in Table I, and the general characteristics of the included RCTs or studies are presented in Table II. The risk of bias assessment for these included RCTs is depicted in Table III. The geometry evidence of the OS network plot and its associated pooled forest plot are summarized in Fig. 2. Furthermore, the geometry evidence of the PFS network plot and its associated pooled forest plot are summarized in Fig. 3. Using SUCRA, graphs of the rank of the treatments associated with OS and PFS are listed in Fig. 4, and the 14 types of treatment-associated toxicities are presented in Fig. 5.

Network diagram (geometry and forest)

Numerous combinations of various treatments were analyzed. The network maps of OS and PFS demonstrated the geometry of 18 chemotherapy regimens compared with a common treatment, Gem, and no loops were identified (Figs. 2A and 3A). Furthermore, the network forest plots indicated the effectiveness of the different regimens compared with the pooled overall result (Figs. 2B and 3B).

Ranking treatments

Due to the variable conditions of APC, a critical aspect to be considered by medical doctors is what chemotherapy regimens are the most suitable and reasonable for the specific conditions of their patients. Therefore, 19 chemotherapy regimens were ranked according to their SUCRA probabilities based on OS and PFS (Fig. 4).

Regarding OS (Fig. 4A), Gem ranked 6th with a SUCRA value of 63.6. The top five combination regimens included Gem+radiotherapy (Radio), Gem+cisplatin (Cis), Gem+erlotinib (Erl)+bevacizumab (Bev), Gem+capecitabine (Cap)+Erl, and Gem+exatecan (Exa). The present results suggested that radiotherapy was the most effective treatment in extending the OS of patients, consistently with the results observed for PFS (Fig. 4B). The SUCRA scores for Gem+irinotecan (Iri) to Gem+tipifarnib presented a similar medium rank, and the scores for Gem+Cap to Gem+vismodegib (Vis) presented a low rank.

For PFS (Fig. 4B), Gem ranked 8th with a SUCRA value of 57.2. The top seven combination regimens included Gem+Radio, Gem+Erl+Bev, Gem+Cis, Gem+Cap+Erl, Gem+pemetrexed, Gem+Iri and Gem+etanercept. The SUCRA scores from Gem+ sorafenib (Sor) to Gem+ nab-paclitaxel (Nab-p) presented a medium rank, and Gem+Cap to Gem+Vis presented a low rank.

Adverse events

In addition to survival, health-associated quality of life issues are a central aspect for patients with APC. Improving the health-associated quality of life by reducing treatment-associated toxicities and the occurrence rate of adverse events is important for patients. The majority of common treatment-associated toxicities include ‘hepatotoxicity’, ‘haematological’, ‘mental/psychiatry’, ‘renal toxicity’, ‘gastrointestinal’, ‘neuropathy’, ‘electrolytes imbalance’, ‘pain’, ‘infection’, ‘skin’, ‘constitutional symptoms’, ‘cardiac/vascular’, ‘pulmonary’ and ‘other’. The majority of these toxicities seriously affect the quality of life of the patient during the treatment process.

Regarding the 19 chemotherapy regimens, each regimen may cause various treatment-associated toxicities, and the occurrence rate of each toxicity varied among treatments. Nevertheless, the three treatment-associated toxicities with the highest occurrence rates were ‘haematological’, ‘gastrointestinal’ and ‘constitutional symptoms’ for all regimens except Gem+Erl, which presented the largest proportions of toxicities. Furthermore, the remaining treatment-associated toxicities, including ‘skin’, ‘hepatotoxicity’, ‘infection’, ‘cardiac/vascular’, ‘neuropathy’, and ‘mental/psychiatry’, were the most common adverse effects among the majority of regimens, following ‘haematological’, ‘gastrointestinal’ and ‘constitutional symptoms’. ‘Renal toxicity’, ‘electrolytes imbalance’ and ‘pulmonary’ presented the lowest occurrence rate among the regimens.

Treatment-associated toxicities always accompany the therapeutic process. To achieve the best results from the perspectives of the clinicians and the patients, patients must consider a series of unavoidable treatment-associated toxicities, leading to a complex selection process.

Discussion

Since Gem was approved as a first-line treatment for APC by the FDA in 1996, a number of combination chemotherapy regimens, including Gem+Xs, have emerged. Post-treatment long-term survival remains poor and is a marked risk of the current chemotherapy regimens (24). This issue may be caused by a failure of local control and of diagnosing localized APC in time (25). Associated RCTs and studies published between 2002 and 2016, covering a total of 14 years, were selected to assess the advantages and disadvantages of each regimen compared with Gem monotherapy.

Among the chemotherapy regimens, Gem+Radio presented the principal improvement in extending OS and PFS. This finding suggested that radiotherapy may block the progressive deterioration associated with advanced cancer and is consistent with the previous study of Youl et al (26), which identified that a gross tumor volume <48 cm3 may be successfully targeted with radiotherapy. An additional three regimens, Gem+Cis, Gem+Erl+Bev and Gem+Cap+Erl, were better compared with Gem monotherapy in terms of OS and PFS. Regarding other combination regimens, Gem+Iri, Gem+Sor, Gem+Erl, Gem+Axitinib, Gem+Exa, Gem+Cetuximab, Gem+Nab-p and Gem+Cap presented higher SUCRA probabilities compared with Gem+5-FU, another double regimen. In contrast, Gem+marismastat and Gem+Vis exhibited decreased SUCRA probabilities. Although numerous regimens are available, side effects always accompany the therapeutic effect. Therefore, selecting the regimen that may offer the longest survival is a complex process that requires the clinician to comprehensively assess the disease status of the patient in order to identify a balance between the benefits and risks of the treatment.

A previous study demonstrated that patients who undergo 5-FU and 5-FU-based regimens following resection had improved survival rates (27). 5-FU has been used as the principle chemoradiation and/or chemotherapy regimens and was previously considered the principal effective chemotherapeutic agent available in pancreatic cancer treatment. In the 1990s, Gem was first demonstrated to be a safe drug with low toxicity for APC treatment (28). Subsequent clinical trials demonstrated the significant advantages of Gem for short-term and long-term OS in the treatment of advanced and metastatic pancreatic cancer (8,29,30); these previous results demonstrated the effectiveness of Gem therapy in pancreatic cancer.

In patients with optimal performance status, concurrent chemotherapy, including FOLFIRINOX, albumin-bound paclitaxel and 5-FU, in combination with Gem, or other Gem-based combined chemotherapies, may provide increased survival benefits (31,32). Although patients at similar disease-stages receive the same chemical regimens, there may be various outcomes due to individual variations.

Gem has been the standard for treating APC since 1996, providing a limited survival of 6 months due to the intrinsic capacity of cancer cells and the surrounding microenvironment to resist cytotoxicity (33,34). Combination therapies with Gem have presented limited effectiveness in clinical trials, and the identification of novel therapeutic strategies that may increase median survival and PFS with reduced adverse effects, is required. Certain treatments, including 5-FU monotherapy, presented significantly decreased survival compared with Gem monotherapy and have demonstrated the effectiveness of Gem as a first-line chemotherapy for APC. Although certain trials examining combination therapies demonstrated improved objective response rates, the treatments failed to achieved improvement in all three of the most common outcomes measured; Median survival, PFS and the objective response rate (3539). The present network meta-analysis of patients with unresectable pancreatic cancer suggested that the current median survival for patients treated with Gem was >6 months, and the objective response rate for Gem ranged between 4.4 and 17.3% (40). These results may represent the standard to be compared with future results of single-arm phase II trials involving Gem-based combination regimens. The severity of the adverse effects among these regimens was assessed by inconsistent scales, which may contribute to bias. Future studies may consider a consistent scale among various conditions in order to avoid possible biases.

In conclusion, numerous chemotherapy regimens are used to extend the survival and reduce the treatment-associated toxicities of patients with APC. The effect and treatment-associated toxicities of a particular regimen requires consideration to balance the benefits and risks for the patient. The present study provides additional evidence for selecting appropriate treatments according to the clinical situation.

Acknowledgements

The authors would like to thank Dr Kun Huang from Chengdu TME Co., Ltd. (Chengdu, China) for advice on optimizing the data search strategy and Professor Jinhui Tian from Lanzhou University (Lanzhou, China) for the valuable discussion regarding the implications of the results.

Funding

The present study was supported by the Science & Technology Department of Sichuan Province Grants (Sichuan, China; grant no. 2016JY0036).

Availability of data and materials

The analyzed data sets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

JC, FX and SY conceived and designed the present study. YX, LC, JY, XW, ZZ and NL performed the data extraction. JY and FX ensured the quality of the data. FX and SY analyzed the data. JC, FX and LC drafted the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Goral V: Pancreatic cancer: Pathogenesis and diagnosis. Asian Pac J Cancer Prev. 16:5619–5624. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Pancreatic Cancer Action Network: The alarming rise of pancreatic cancer deaths in the United States. Why we need to stem the tide today. Pancreatic Cancer Action Network; Manhattan Beach, CA: 2012

3 

Krechler T, Horejs J, Ulrych J, Zeman M, Macásek J, Dusková J and Zák A: Current status of pancreatic cancer diagnosis. Cas Lek Cesk. 150:587–593. 2011.(In Czech). PubMed/NCBI

4 

Freelove R and Walling AD: Pancreatic cancer: Diagnosis and management. Am Fam Physician. 73:485–492. 2006.PubMed/NCBI

5 

Chen WQ, Liang D, Zhang SW, Zheng RS and He YT: Pancreatic cancer incidence and mortality patterns in china, 2009. Asian Pac J Cancer Prev. 14:7321–7324. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Wang Y, Schrag D, Brooks GA and Dominici F: National trends in pancreatic cancer outcomes and pattern of care among Medicare beneficiaries, 2000 through 2010. Cancer. 120:1050–1058. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Akhtar-Danesh GG, Finley C and Akhtar-Danesh N: Long-term trends in the incidence and relative survival of pancreatic cancer in Canada: A population-based study. Pancreatology. 16:259–265. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Burris HA III, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P, et al: Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J Clin Oncol. 15:2403–2413. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Abbruzzese JL: New applications of gemcitabine and future directions in the management of pancreatic cancer. Cancer. 95 (Suppl 4):941–945. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Macdonald JS: Clinical overview: Adjuvant therapy of gastrointestinal cancer. Cancer Chemother Pharmacol. 54 Suppl 1:S4–S11. 2004.PubMed/NCBI

11 

Okusaka T, Ishii H, Funakoshi A, Ueno H, Furuse J and Sumii T: A phase I/II study of combination chemotherapy with gemcitabine and 5-fluorouracil for advanced pancreatic cancer. Jpn J Clin Oncol. 36:557–563. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Shi S, Yao W, Xu J, Long J, Liu C and Yu X: Combinational therapy: New hope for pancreatic cancer? Cancer Lett. 317:127–135. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Kristensen A, Vagnildhaug OM, Grønberg BH, Kaasa S, Laird B and Solheim TS: Does chemotherapy improve health-related quality of life in advanced pancreatic cancer? A systematic review. Crit Rev Oncol Hematol. 99:286–298. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Gresham GK, Wells GA, Gill S, Cameron C and Jonker DJ: Chemotherapy regimens for advanced pancreatic cancer: A systematic review and network meta-analysis. BMC Cancer. 14:4712014. View Article : Google Scholar : PubMed/NCBI

15 

Ansari D, Tingstedt B, Andersson B, Holmquist F, Sturesson C, Williamsson C, Sasor A, Borg D, Bauden M and Andersson R: Pancreatic cancer: Yesterday, today and tomorrow. Future Oncol. 12:1929–1946. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Birhanu G, Javar HA, Seyedjafari E and Zandi-Karimi A: Nanotechnology for delivery of gemcitabine to treat pancreatic cancer. Biomed Pharmacother. 88:635–643. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Moher D, Liberati A, Tetzlaff J and Altman DG: PRISMA Group: Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 6:e10000972009. View Article : Google Scholar : PubMed/NCBI

19 

Chaimani A, Higgins JP, Mavridis D, Spyridonos P and Salanti G: Graphical tools for network meta-analysis in STATA. PLoS One. 8:e766542013. View Article : Google Scholar : PubMed/NCBI

20 

Biermann JS, Chow W, Reed DR, Lucas D, Adkins DR, Agulnik M, Benjamin RS, Brigman B, Budd GT, Curry WT, et al: NCCN guidelines insights: Bone cancer, version 2.2017. J Natl Compr Canc Netw. 15:155–167. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Hernández-Quiles C, Bernabeu-Wittel M, Pérez-Belmonte LM, Macías-Mir P, Camacho-González D, Massa B, Maiz-Jiménez M and Ollero-Baturone M: PALIAR investigators: Concordance of Barthel Index, ECOG-PS, and Palliative Performance Scale in the assessment of functional status in patients with advanced medical diseases. BMJ Support Palliat Care. 7:300–307. 2017.PubMed/NCBI

22 

Lovato N, Lack L, Wright H and Kennaway DJ: Evaluation of a brief treatment program of cognitive behavior therapy for insomnia in older adults. Sleep. 37:117–126. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Salanti G, Ades AE and Ioannidis JP: Graphical methods and numerical summaries for presenting results from multiple-treatment meta-analysis: An overview and tutorial. J Clin Epidemiol. 64:163–171. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Jutric Z and Melstrom LG: New treatment options and management considerations in borderline resectable pancreatic cancer. Oncology (Williston Park). 31:443–452. 2017.PubMed/NCBI

25 

Sugarbaker PH: Strategies to improve local control of resected pancreas adenocarcinoma. Surg Oncol. 26:63–70. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Youl M, Hashem S, Brade A, Cummings B, Dawson LA, Gallinger S, Hedley D, Jiang H, Kim J, Krzyzanowska MK, et al: Induction gemcitabine plus concurrent gemcitabine and radiotherapy for locally advanced unresectable or resected pancreatic cancer. Clin Oncol (R Coll Radiol). 26:203–209. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Kalser MH and Ellenberg SS: Pancreatic cancer. Adjuvant combined radiation and chemotherapy following curative resection. Arch Surg. 120:899–903. 1985. View Article : Google Scholar : PubMed/NCBI

28 

Aapro MS, Martin C and Hatty S: Gemcitabine-a safety review. Anticancer Drugs. 9:191–201. 1998. View Article : Google Scholar : PubMed/NCBI

29 

Regine WF, Winter KA, Abrams RA, Safran H, Hoffman JP, Konski A, Benson AB, Macdonald JS, Kudrimoti MR, Fromm ML, et al: Fluorouracil vs gemcitabine chemotherapy before and after fluorouracil-based chemoradiation following resection of pancreatic adenocarcinoma: A randomized controlled trial. JAMA. 299:1019–1026. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Cunningham D, Chau I, Stocken DD, Valle JW, Smith D, Steward W, Harper PG, Dunn J, Tudur-Smith C, West J, et al: Phase III randomized comparison of gemcitabine versus gemcitabine plus capecitabine in patients with advanced pancreatic cancer. J Clin Oncol. 27:5513–5518. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Vaccaro V, Sperduti I and Milella M: FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 365:768–769; author reply 769. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C, et al: FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 364:1817–1825. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, et al: Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 25:735–747. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Fox RG, Lytle NK, Jaquish DV, Park FD, Ito T, Bajaj J, Koechlein CS, Zimdahl B, Yano M, Kopp J, et al: Image-based detection and targeting of therapy resistance in pancreatic adenocarcinoma. Nature. 534:407–411. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Citterio C, Baccini M, Orlandi E, Di Nunzio C and Cavanna L: Second-line chemotherapy for the treatment of metastatic pancreatic cancer after first-line gemcitabine-based chemotherapy: A network meta-analysis. Oncotarget. 9:29801–29809. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Chiorean EG, Von Hoff DD, Tabernero J, El-Maraghi R, Wee Ma W, Reni M, Harris M, Whorf R, Liu H, Shiansong Li J, et al: Second-line therapy after nab-paclitaxel plus gemcitabine or after gemcitabine for patients with metastatic pancreatic cancer. Br J Cancer. 115:e132016. View Article : Google Scholar : PubMed/NCBI

37 

Oettle H, Riess H, Stieler JM, Heil G, Schwaner I, Seraphin J, Görner M, Mölle M, Greten TF, Lakner V, et al: Second-line oxaliplatin, folinic acid, and fluorouracil versus folinic acid and fluorouracil alone for gemcitabine-refractory pancreatic cancer: Outcomes from the CONKO-003 trial. J Clin Oncol. 32:2423–2429. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Walker EJ and Ko AH: Beyond first-line chemotherapy for advanced pancreatic cancer: An expanding array of therapeutic options? World J Gastroenterol. 20:2224–2236. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Schultheis B, Reuter D, Ebert MP, Siveke J, Kerkhoff A, Berdel WE, Hofheinz R, Behringer DM, Schmidt WE, Goker E, et al: Gemcitabine combined with the monoclonal antibody nimotuzumab is an active first-line regimen in KRAS wildtype patients with locally advanced or metastatic pancreatic cancer: A multicenter, randomized phase IIb study. Ann Oncol. 28:2429–2435. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Colucci G, Giuliani F, Gebbia V, Biglietto M, Rabitti P, Uomo G, Cigolari S, Testa A, Maiello E and Lopez M: Gemcitabine alone or with cisplatin for the treatment of patients with locally advanced and/or metastatic pancreatic carcinoma: A prospective, randomized phase III study of the Gruppo Oncologia dell'Italia Meridionale. Cancer. 94:902–910. 2002. View Article : Google Scholar : PubMed/NCBI

41 

Berlin JD, Catalano P, Thomas JP, Kugler JW, Haller DG and Benson AB III: 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. 20:3270–3275. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Bramhall SR, Schulz J, Nemunaitis J, Brown PD, Baillet M and Buckels JA: A double-blind placebo-controlled, randomised study comparing gemcitabine and marimastat with gemcitabine and placebo as first line therapy in patients with advanced pancreatic cancer. Br J Cancer. 87:161–167. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Rocha Lima CM, Green MR, Rotche R, Miller WH Jr, Jeffrey GM, Cisar LA, Morganti A, Orlando N, Gruia G and Miller LL: Irinotecan plus gemcitabine results in no survival advantage compared with gemcitabine monotherapy in patients with locally advanced or metastatic pancreatic cancer despite increased tumor response rate. J Clin Oncol. 22:3776–3783. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Van Cutsem E, van de Velde H, Karasek P, Oettle H, Vervenne WL, Szawlowski A, Schoffski P, Post S, Verslype C, Neumann H, et al: Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol. 22:1430–1438. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Oettle H, Richards D, Ramanathan RK, van Laethem JL, Peeters M, Fuchs M, Zimmermann A, John W, Von Hoff D, Arning M and Kindler HL: A phase III trial of pemetrexed plus gemcitabine versus gemcitabine in patients with unresectable or metastatic pancreatic cancer. Ann Oncol. 16:1639–1645. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Von Hoff DD, Ervin TJ, Arena FP, Gabriela Chiorean E, Infante JR, Moore M, Seay TE, Tjulandin S, Ma WW, Saleh M, et al: Randomized phase III study of weekly nab-paclitaxel plus gemcitabine versus gemcitabine alone in patients with metastatic adenocarcinoma of the pancreas (MPACT). J Clin Oncol. 31:LBA148. 2013. View Article : Google Scholar

47 

Abou-Alfa GK, Letourneau R, Harker G, Modiano M, Hurwitz H, Tchekmedyian NS, Feit K, Ackerman J, De Jager RL, Eckhardt SG and O'Reilly EM: Randomized phase III study of exatecan and gemcitabine compared with gemcitabine alone in untreated advanced pancreatic cancer. J Clin Oncol. 24:4441–4447. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Heinemann V, Quietzsch D, Gieseler F, Gonnermann M, Schönekäs H, Rost A, Neuhaus H, Haag C, Clemens M, Heinrich B, et al: Randomized phase III trial of gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer. J Clin Oncol. 24:3946–3952. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Stathopoulos GP, Syrigos K, Aravantinos G, Polyzos A, Papakotoulas P, Fountzilas G, Potamianou A, Ziras N, Boukovinas J, Varthalitis J, et al: A multicenter phase III trial comparing irinotecan-gemcitabine (IG) with gemcitabine (G) monotherapy as first-line treatment in patients with locally advanced or metastatic pancreatic cancer. Br J Cancer. 95:587–592. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Herrmann R, Bodoky G, Ruhstaller T, Glimelius B, Bajetta E, Schüller J, Saletti P, Bauer J, Figer A, Pestalozzi B, 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. 25:2212–2217. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, 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. 25:1960–1966. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Spano JP, Chodkiewicz C, Maurel J, Wong R, Wasan H, Barone C, Létourneau R, Bajetta E, Pithavala Y, Bycott P, et al: Efficacy of gemcitabine plus axitinib compared with gemcitabine alone in patients with advanced pancreatic cancer: An open-label randomised phase II study. Lancet. 371:2101–2108. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Philip PA, Benedetti J, Corless CL, Wong R, O'Reilly EM, Flynn PJ, Rowland KM, Atkins JN, Mirtsching BC, Rivkin SE, et al: Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. J Clin Oncol. 28:3605–3610. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Kindler HL, Ioka T, Richel DJ, Bennouna J, Létourneau R, Okusaka T, Funakoshi A, Furuse J, Park YS, Ohkawa S, et al: Axitinib plus gemcitabine versus placebo plus gemcitabine in patients with advanced pancreatic adenocarcinoma: A double-blind randomised phase 3 study. Lancet Oncol. 12:256–262. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Loehrer PJ Sr, Feng Y, Cardenes H, Wagner L, Brell JM, Cella D, Flynn P, Ramanathan RK, Crane CH, Alberts SR and Benson AB III: Gemcitabine alone versus gemcitabine plus radiotherapy in patients with locally advanced pancreatic cancer: An Eastern Cooperative Oncology Group trial. J Clin Oncol. 29:4105–4112. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Heinemann V, Vehling-Kaiser U, Waldschmidt D, Kettner E, Märten A, Winkelmann C, Klein S, Kojouharoff G, Gauler TC, von Weikersthal LF, et al: Gemcitabine plus erlotinib followed by capecitabine versus capecitabine plus erlotinib followed by gemcitabine in advanced pancreatic cancer: Final results of a randomised phase 3 trial of the ‘Arbeitsgemeinschaft Internistische Onkologie’ (AIO-PK0104). Gut. 62:751–759. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Wu C, Fernandez SA, Criswell T, Chidiac TA, Guttridge D, Villalona-Calero M and Bekaii-Saab TS: Disrupting cytokine signaling in pancreatic cancer: A phase I/II study of etanercept in combination with gemcitabine in patients with advanced disease. Pancreas. 42:813–818. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Moehler M, Maderer A, Schimanski C, Kanzler S, Denzer U, Kolligs FT, Ebert MP, Distelrath A, Geissler M, Trojan J, et al: Gemcitabine plus sorafenib versus gemcitabine alone in advanced biliary tract cancer: A double-blind placebo-controlled multicentre phase II AIO study with biomarker and serum programme. Eur J Cancer. 50:3125–3135. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Catenacci DV, Junttila MR, Karrison T, Bahary N, Horiba MN, Nattam SR, Marsh R, Wallace J, Kozloff M, Rajdev L, et al: Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J Clin Oncol. 33:4284–4292. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Ramanathan RK, Goldstein D, Korn RL, Arena F, Moore M, Siena S, Teixeira L, Tabernero J, Van Laethem JL, Liu H, et al: Positron emission tomography response evaluation from a randomized phase III trial of weekly nab-paclitaxel plus gemcitabine versus gemcitabine alone for patients with metastatic adenocarcinoma of the pancreas. Ann Oncol. 27:648–653. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 19 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen J, Chen L, Yu J, Xu Y, Wang X, Zeng Z, Liu N, Xu F and Yang S: Meta‑analysis of current chemotherapy regimens in advanced pancreatic cancer to prolong survival and reduce treatment‑associated toxicities. Mol Med Rep 19: 477-489, 2019
APA
Chen, J., Chen, L., Yu, J., Xu, Y., Wang, X., Zeng, Z. ... Yang, S. (2019). Meta‑analysis of current chemotherapy regimens in advanced pancreatic cancer to prolong survival and reduce treatment‑associated toxicities. Molecular Medicine Reports, 19, 477-489. https://doi.org/10.3892/mmr.2018.9638
MLA
Chen, J., Chen, L., Yu, J., Xu, Y., Wang, X., Zeng, Z., Liu, N., Xu, F., Yang, S."Meta‑analysis of current chemotherapy regimens in advanced pancreatic cancer to prolong survival and reduce treatment‑associated toxicities". Molecular Medicine Reports 19.1 (2019): 477-489.
Chicago
Chen, J., Chen, L., Yu, J., Xu, Y., Wang, X., Zeng, Z., Liu, N., Xu, F., Yang, S."Meta‑analysis of current chemotherapy regimens in advanced pancreatic cancer to prolong survival and reduce treatment‑associated toxicities". Molecular Medicine Reports 19, no. 1 (2019): 477-489. https://doi.org/10.3892/mmr.2018.9638