Keywords: gastric cancer; ramucirumab; paclitaxel; second line therapy; vascular endothelial growth factor receptor 2; targeted therapy; nutrition

Similar documents
Chemotherapy for Advanced Gastric Cancer

pan-canadian Oncology Drug Review Final Clinical Guidance Report Ramucirumab (Cyramza) for Gastric Cancer October 29, 2015

Targeted Therapies in Metastatic Colorectal Cancer: An Update

Gastric cancer and lung cancer impose a substantial

ESMO 2017, Madrid, Spain Dr. Loredana Vecchione Charite Comprehensive Cancer Center, Berlin HIGHLIGHTS ON CANCERS OF THE UPPER GI TRACT

Cancer Cell Research 14 (2017)

Gastric cancer is the fourth

Bodoky G, Gil-Delgado M, Cascinu S, Lipatov ON, Cunningham D, Van Cutsem E, Muro K, Chandrawansa K, Liepa AM, Carlesi R, Ohtsu A, Wilke H

Van Cutsem E et al. Proc ASCO 2009;Abstract LBA4509.

Updated Apr 2017 by Dr. Ko (Medical Oncologist, Abbotsford Cancer Centre)

Highlights STOMACH CANCER

A retrospective study of the safety and efficacy of paclitaxel plus ramucirumab in patients with advanced or recurrent gastric cancer with ascites

ONCOLOGY LETTERS 2: , 2011

GASTRIC & PANCREATIC CANCER

Conflicts of Interest GI Malignancies: An Update on Current Treatment Options

Analysis of esophagogastric cancer patients enrolled in the National Cancer Institute Cancer Therapy Evaluation Program sponsored phase 1 trials

General Information, efficacy and safety data

Advances in gastric cancer: How to approach localised disease?

This clinical study synopsis is provided in line with Boehringer Ingelheim s Policy on Transparency and Publication of Clinical Study Data.

Medicinae Doctoris. One university. Many futures.

Case 1 Metastatic Pancreatic Adenocarcinoma: What Therapy Should I Select First?

National Horizon Scanning Centre. Bevacizumab (Avastin) in combination with non-taxanes for metastatic breast cancer - first line therapy

MEETING SUMMARY ESMO 2018, Munich, Germany. Dr. Jenny Seligmann University of Leeds, UK HIGHLIGHTS ON COLORECTAL CANCER

ASCO 2017 updates in Colorectal and Gastric Cancers. May Cho, M.D.

Gemcitabine and Carboplatin in Patients with Refractory or Progressive Metastatic Breast Cancer after Treatment

Technology appraisal guidance Published: 27 January 2016 nice.org.uk/guidance/ta378

Karcinom dojke. PANEL: Semir Bešlija, Zdenka Gojković, Robert Šeparović, Tajana Silovski

Novel Chemotherapy Agents for Metastatic Breast Cancer. Joanne L. Blum, MD, PhD Baylor-Sammons Cancer Center Dallas, TX

New chemotherapy drugs in metastatic breast cancer. Guy Jerusalem, MD, PhD

Edith A. Perez, Ahmad Awada, Joyce O Shaughnessy, Hope Rugo, Chris Twelves, Seock-Ah Im, Carol Zhao, Ute Hoch, Alison L. Hannah, Javier Cortes

Materials and Methods

Sponsor / Company: Sanofi Drug substance(s): Docetaxel (Taxotere )

DALLA CAPECITABINA AL TAS 102

Tegafur, gimeracil, and oteracil (known as S1) for first-line palliative treatment of advanced gastric cancer

IMMUNOTHERAPY FOR GASTROINTESTINAL CANCERS

Antiangiogenic therapy in GI cancer: current status and future directions

Targeted Agents as Maintenance Therapy. Karen Kelly, MD Professor of Medicine UC Davis Cancer Center

Getting to the Bottom of Treatment: An Update in the Management of Esophagogastric Cancers

OUR EXPERIENCES WITH ERLOTINIB IN SECOND AND THIRD LINE TREATMENT PATIENTS WITH ADVANCED STAGE IIIB/ IV NON-SMALL CELL LUNG CANCER

Neo- and adjuvant treatment for gastric cancer: The role of chemotherapy

Cetuximab plus 5-FU/FA/oxaliplatin (FOLFOX-4) in the first-line treatment of metastatic colorectal cancer: a large-scale Phase II study (OPUS)

Updates and best practices in the management of gastric cancer

Recent advances in the management of metastatic breast cancer in older adults

Evolving Paradigms in HER2+ MBC: Strategies for Individualizing Therapy with Available Agents

Published Ahead of Print on August 20, 2018 as /theoncologist

New Targeted Agents Demonstrate Greater Efficacy and Tolerability in the Treatment of HER2-positive Breast Cancer

The legally binding text is the original French version

CHEMOTHERAPY FOR METASTATIC GASTRIC CANCER

DR LUIS MANSO UNIDAD TUMORES DE MAMA Y GINECOLÓGICOS HOSPITAL 12 DE OCTUBRE MADRID

This clinical study synopsis is provided in line with Boehringer Ingelheim s Policy on Transparency and Publication of Clinical Study Data.

The following slides are provided as presented by the author during the live educa7onal ac7vity and are intended for reference purposes only.

Management Guidelines and Targeted Therapies in Metastatic Non-Small Cell Lung Cancer: An Oncologist s Perspective

trial update clinical

The next wave of successful drug therapy strategies in HER2-positive breast cancer. Hans Wildiers University Hospitals Leuven Belgium

Jonathan Dickinson, LCL Xeloda

METRIC Study Key Eligibility Criteria

Summary Background 1. Comparative effectiveness of ramucirumab

Common disease 175,000 new cases/year 44,000 deaths/year Less than 10% with newly diagnosed at presentation have stage IV disease Chronic disease,

Nivolumab: esperienze italiane nel carcinoma polmonare avanzato

Adjuvant Chemotherapy

Maintenance paradigm in non-squamous NSCLC

INMUNOTERAPIA I. Dra. Virginia Calvo

Upper Gastrointestinal Cancers in the Elderly. Choo Su Pin Senior Consultant Medical Oncology National Cancer Centre Singapore

Locoregional treatment Session Oral Abstract Presentation Saulo Brito Silva

First-line single-agent chemotherapy for patients with recurrent or metastatic gastric cancer with poor performance status

Possible causes of difference among regions How to look at the results from MRCT Non-compliance with GCP and/or protocol Apparent Differences (Play of

MEETING SUMMARY ASCO GI, SATURDAY JANUARY 17 TH 2015

NOVITA IN TEMA DI CARCINOMA GASTRICO ROSA BERENATO

National Horizon Scanning Centre. Bevacizumab (Avastin) for glioblastoma multiforme - relapsed. August 2008

Summary of the study protocol of the FLOT3-Study

Avastin NAME OF THE MEDICINE DESCRIPTION PHARMACOLOGY. bevacizumab (rch)

Cancer du sein métastatique et amélioration de la survie Pr. X. Pivot

Research Article Prognostic Factors in Advanced Non-Small-Cell Lung Cancer Patients: Patient Characteristics and Type of Chemotherapy

Low Dose Docetaxel Combined With Low Dose Capecitabine in Treatment of Metastatic Breast Cancer Previously Treated With Anthracycline

Now Available: Final Rule for FDAAA 801 and NIH Policy on Clinical Trial Reporting

Cisplatin plus Gemcitabine versus Gemcitabine for Biliary Tract Cancer. Valle J et al. N Engl J Med 2010;362(14):

STUDY FINDINGS PRESENTED ON TAXOTERE REGIMENS IN HEAD AND NECK, LUNG AND BREAST CANCER

Immune checkpoint blockade in lung cancer

BIOLOGICAL TARGETED AGENTS

EGFR inhibitors in NSCLC

Update on the Management of HER2+ Breast Cancer. Christian Jackisch, MD, PhD Sana Klinikum Offenbach Offenbach, Germany

Ca Cardias e Stomaco: le diversita e le terapie

FoROMe Lausanne 6 février Anita Wolfer MD-PhD Cheffe de clinique Département d Oncologie, CHUV

Immunotherapy for Breast Cancer. Aurelio B. Castrellon Medical Oncology Memorial Healthcare System

Incorporating biologics in the management of older patients with metastatic colorectal cancer

Sponsor / Company: Sanofi Drug substance(s): SAR (iniparib)

Combining Lurbinectedin and Doxorubicin The UCLH Experience in Small Cell Lung Cancer

Systemic treatment in early and advanced gastric cancer

Role of Primary Resection for Patients with Oligometastatic Disease

1 st Appraisal Committee meeting Background & Clinical Effectiveness Gillian Ells & Malcolm Oswald 24/11/2016

Breast Cancer Immunotherapy. Leisha A. Emens, MD PhD Johns Hopkins University Bloomberg Kimmel Institute for Cancer Immunotherapy

Evaluation of pharmacological therapies used in Costa Rica in patients with metastatic gastric cancer: a retrospective study

Bevacizumab for the treatment of recurrent advanced ovarian cancer

Docetaxel. Class: Antineoplastic agent, Antimicrotubular, Taxane derivative.

Thoracic and head/neck oncology new developments

Breast cancer treatment

Metastatic NSCLC: Expanding Role of Immunotherapy. Evan W. Alley, MD, PhD Abramson Cancer Center at Penn Presbyterian

GAP (Gemcitabine Abraxane Pancreas) Trial. Codice Eudract Sponsor non profit: Rossana Berardi, MD Alessandro Bittoni, MD

THE ESMO-MAGNITUDE OF CLINICAL BENEFIT SCALE V1.1

Transcription:

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 Original article THE IMPACT OF CLINICAL FEATURES ON EFFICACY AND SAFETY OF THE COMBINATION OF RAMUCIRUMAB AND PACLITAXEL FOR METASTATIC GASTRO-OESOPHAGEAL JUNCTION/ GASTRIC CANCER: DATA OF A REAL LIFE EXPERIENCE IN AN ITALIAN INSTITUTION. Emanuele Vita 1*, Antonia Strippoli 1*, Vincenzo Di Noia 1, Carmela Di Dio 1, Valeria Zurlo 1, Michele Basso 1, Ettore D Argento 1, Schinzari Giovanni 1, Carlo Barone 1, Alessandra Cassano 1 1 Department of Medical Oncology, Fondazione Policlinico A Gemelli Università Cattolica del Sacro Cuore, Rome, Italy; email: dh.oncologia@policlinicogemelli.it Correspondence: dr.emanuele.vita@gmail.com; strefoantonia@gmail.com Tel. +39 3480510228 + 39 0630156318 * Reference author: Dr. Emanuele Vita, Dr.ssa Antonia Strippoli. Both authors contributed equally to this work Abstract: The RAINBOW Phase III study established the efficacy of the combination of paclitaxel and ramucirumab, a monoclonal antibody targeting VEGF receptor-2 (VEGF-R2), as second-line therapy. We retrospectively analyzed the data of patients treated with ramucirumab plus paclitaxel at our Institution to evaluate the impact of clinical heterogeneous figures on the efficacy and safety of this combination paclitaxel/ramucirumab in a real- life cohort of patients. After a median followup of 10.74 months, the median progression-free survival (PFS) was 5.8 months (95% CI: 3.04-5,63). Disease control rate (DCR) was 61% and the median duration of response (DOR) was 5.8 months. Median overall survival (OS) was 8.3 months. A trend toward better outcome was observed in HER2 positive patients. In multivariate analysis, nutritional status (p = 0.0001) and number of metastatic sites (p = 0.0266) resulted significantly related with longer PFS. Our analysis confirmed the efficacy and safety of the combination of ramucirumab with paclitaxel also in the real-life practice and the median PFS is significantly longer than that reported for Western population in previous studies. Subgroup analysis confirms the key-role of nutritional status as prognostic factor and suggests a possible interaction between EGF and angiogenesis pathways that deserves further investigations. Keywords: gastric cancer; ramucirumab; paclitaxel; second line therapy; vascular endothelial growth factor receptor 2; targeted therapy; nutrition 1. Introduction Gastric cancer is a leading cause of cancer mortality worldwide, ranking the fourth in Italy, where it was responsible for 9557 deaths in 2014 [1, 2]. In 2017, 12800 new diagnoses of gastrooesophageal junction and gastric cancer have been estimated in Italy; most patients are diagnosed in advanced/metastatic stage or develop a relapse after surgery with curative intent. A combination of platinum compounds (oxaliplatin and cisplatin) and fluoropyrimidines (fluorouracil, capecitabine, and S1) is generally considered as the standard-of-care for first line treatment; targeted therapy is limited to HER-2 positive disease that is treated with trastuzumab, in addition to chemotherapy [3]. For patient who progressed after first-line CT and have good clinical conditions, the association of paclitaxel and ramucirumab, a monoclonal antibody targeting VEGF receptor-2 (VEGF-R2), is currently the preferred therapy, based on the result of the RAINBOW trial that showed significant survival benefits in comparison to chemotherapy alone. Moreover, in the REGARD trial ramucirumab alone has produced a statistically significant improvement of life expectancy over placebo in second line setting, proving to be a useful option for patient who are unfit for chemotherapy [4,5]. 2018 by the author(s). Distributed under a Creative Commons CC BY license.

2 of 13 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 On the basis of this background, we report our experience with the combination of PACLITAXEL and RAMUCIRUMAB in pre-treated metastatic gastric (mgc) ad gastro-oesophageal cancer (mgoc), in order to assess the safety and efficacy of this regimen in the daily clinical practice. 2. Results 2.1 Patients characteristics All patients received at least 1 cycle (28 days) of the drug combination and have at least a 2 months follow-up. The longest treatment period was 15 cycles and it is still ongoing. The characteristics of patients and their tumors are summarized in table 1. The primary tumor site was stomach in 34 patients (72,3 %) or gastro-oesophageal junction in 13 patients (27,7 %), respectively. The histology subtype according to Lauren classification was intestinal or adenocarcinoma NOS in 33 cases and diffuse with signet ring cells in 14 cases. Primary tumor was previously resected in twenty-five patients, including five cases of palliative surgery due to bleeding, and twenty cases of radical surgery, associated with neoadjuvant/perioperative therapy (5 cases), intraoperative HIPEC (1 case) or adjuvant therapy (10 cases). Forty-four patients were previously treated with platinum and fluoropyrimidine doublets, including twelve cases of HER-2 positive tumors who received a combination regimen with trastuzumab according to the approved recommendations; only three patients had received triplet regimens with platinum, fluoropyrimidine and anthracycline. No patient was previously exposed to taxanes, whereas seven patients were treated with paclitaxel and ramucirumab after failure of a previous second-line therapy with FOLFIRI. Several patients had poor prognostic characteristics: twenty-one patients (44.6%) experienced disease recurrence during adjuvant therapy (2) or had disease progression within 6 months from the start of first-line therapy (19); 20 patients (42,7 %) had peritoneal carcinosis and 12 patients (25,5%) had at least three metastatic sites, including brain metastases (2 cases); 1/3 of patients were underweight as expression of nutritional deficiency and low meal intake. After failure of therapy with paclitaxel and ramucirumab, thirteen patients received a subsequent line of therapy, mainly an irinotecan-based regimen (10 cases).

Preprints (www.preprints.org) NOT PEER-REVIEWED Posted: 24 April 2018 3 of 13 Table 1. Patients' characteristics Age (range): median 56 years (32-78) Site of primary tumour <65 33 (70,2 %) Gastric adenocarcinoma 34 (72,3 %) 65 14 (19,8 %) GEJ adenocarcinoma 13 (27,7 %) Sex ECOG performance status Male 29 (61,7 %) (median age 59 years; 44-76) ECOG 0 29 (61,7 %) Female 18 (38,3 %) (median age 50 years; 32-78) ECOG 1 18 (38,3 %) Histological subtype (Lauren classification) HER-2 status Intestinal/adenocarcinoma NAS 33 (70,2 %) Positive 12 (25,5 %) Diffuse 14 (19,8 %) Negative 35 (74,5 %) Number of metastatic sites Peritoneal metastases 0-2 35 (74,5 %) YES 20 (42,5 %) 3 NO 27 (57,5 %) 12 (25,5 %) TTP on previous platinum-based therapy <6 months 21 (44,6 %) 94 95 96 97 98 99 100 101 102 103 104 Previous platinum exposition Oxaliplaltin 12 (25,5 %) 6 months 26 (55,4 %) Cisplatin 35 (74,5 %) Previous treatment with FOLFIRI Subsequent line with FOLFIRI YES 7 (14,9 %) YES 10 (21,3 %) NO 40 (85,1 %) NO 37 (78,7 %) Previous surgery for gastric cancer Nutritional status YES 25 (53,1 %) Normal weight (BMI 15) 34 (72,4 %) NO 22 (46,9 %) Underweight (BMI <15) 13 (27,6 %) 2.2 Efficacy analysis As of data cutoff (March 31, 2018), after a median follow-up of 10.74 months, 31 patients have experienced disease progression, while 16 patients are still on treatment. Median PFS was 5.4 months (95% CI: 3.04 5.63) (fig. 1). Objective radiological response to treatment is reported in table 2. Among forty-one evaluable patients, we observed ten partial responses, including two complete responses according to mrecist criteria, and fifteen stable disease, accounting for a disease control rate of 61%; three partial responses and one stable disease were achieved in patients who had previously received two lines of therapy. The median duration of response was 5.8 months (95% CI: 5.02 6.80).

4 of 13 124 125 126 127 Table 2. Best overall response Response N (%) (41 patients) Complete response 0 (0%) Partial response 10 (24.4%) Stable disease 15 (36,6%) Disease control rate (CR + PR + SD) 25 (61 %) Progressive disease 16 (39%) 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 At the time of the analysis, 23 patients had died. Early deaths (within 30 days of last treatment) were observed in three patients and were related to clinical disease progression. The median OS was 8.3 months (95% CI: 6.2 11.0) (fig. 1).

5 of 13 128 129 130 131 132 133 134 135 136 137 Figure 1. Kaplan-Maier curves for PFS and OS. Results of the univariate analyses for clinical characteristics are summarized in table 3. Despite no statistically significant difference emerged, it is interesting to note that the better outcomes, including longer PFS (median PFS 6.8 months) and higher disease control rate (8 out of 9 evaluable patients, accounting for RR 44% and DCR 89%), have been recorded in HER-2 positive patients, that also resulted the subgroup with higher number of ongoing treatments (6/12, 50%) and the shortest median time of follow-up (8.64 months). Neither factors associated with previous treatments (TTP after first line, platinum compound used,

6 of 13 138 139 140 exposure to irinotecan, gastrectomy) nor the presence of peritoneal metastases had a significant impact on survival outcomes. Table 3. Subgroups analysis for PFS and OS PFS median HR (CI 95%) OS median HR (CI 95%) Age (< 65 vs 65) 5.27 mo. vs 7.23 mo. HR 0.57 (0.23 1.41) p = 0.2088 7.8 mo. vs 10.45 mo. HR 0.57 (0.19 1.69) p= 0.3192 Gender (Male vs Female) 5.7 mo. vs 3.83 mo. HR 1.14 (0.55 2.38) p = 0.7151 7.8 mo. vs 8.3 mo HR 1.00 (0.2 2. 36) p = 0.9958 Histology (diffuse vs intestinal/adc NAS) 4.62 mo. vs 5.6 mo. HR 1.02 (0.50-2.10) p = 0.9377 7.8 mo. vs 8.3 mo. HR 0.91 (0.40 2.09) p = 0.8362 HER 2 status (positive vs negative) 6.8 mo. vs 5.27 mo. HR 0.51 (0.25 1.19) p = 0.1341 10.25 vs 7.8 mo. HR 0.65 (0.25 1.78) p = 0.4364 Site of primary tumor (EGJ vs stomach) 5.7 mo. vs 5.27 mo. HR 1.15 (0.51 2.59) p =0.7237 7.8 mo. vs. 11.4 mo. HR 1.93 (0.65 5.7) p = 0.2350 Previous gastrectomy (NO vs YES) 4.13 mo. vs 5.9 mo. HR 0.55 (0.27 1.11) p = 0.1013 6.73 mo. vs 11.40 mo. HR 0.50 (0.21 1.17) p = 0.1132 Metastatic sites (1-2 vs 3) 5.7 mo. vs 2.77 mo. HR 0.42 (0.20 0.92) p = 0.0266 10.45 mo. vs 5.63 mo. HR 0.39 (0.10 0.83) p = 0.0212 Peritoneal metastases (YES vs NO) 3.83 mo. vs 5.9 mo. HR 0.56 (0.27 1.15) p = 0.1184 7.5 mo. vs 10.45 mo. HR 2.39 (1.00 5.7) p = 0.0490 PS ECOG (0 vs 1) 5.7 mo. vs 3.97 mo. HR 0.60 (0.29-1.23) p = 0.1584 11.4 mo. vs 6.0 mo. HR 0.29 (HR 0.08 0.56) P = 0.0016 Nutritional status (BMI 15 vs BMI < 15) 6.5 mo. vs 2.57 mo. HR 0.17 (0.07 0.41) p = 0.0001 10.45 mo. 4.73 mo. HR 0.29 (0.07 0.54) p = 0.0016 PFS with first line CT (> 6 mo. vs < 6 mo.) 5.67 mo. vs. 3.46 mo. 11.4 mo. vs 6.7 mo.

7 of 13 141 142 143 144 145 146 147 148 149 150 Previous platinum-based CT (OXA vs CDDP) Previous FOLFIRI treatment (NO vs YES) HR 0.49 (0.21 0.97) p = 0.0431 3.63 mo. vs 5.9 mo. HR 0.36 (1.40 10.23) p = 0.0114 5.27 mo. vs 7.9 mo. HR 0.51 (0.17 1.47) p = 0.2191 HR 0.36 (0.14 0.79) p = 0.0126 7.5 mo. vs 9.1 mo. HR 1.38 (0.54 3.82) p = 0.4586 7.8 mo. vs 11.4 mo. HR 2.11 (0.59 5.54) p = 0.2904 In the multivariable stepwise Cox regression analysis only three factors resulted associated with poor prognosis: high number of metastatic sites (more than two), BMI < 15 and PS ECOG of 1 (table 4). Patients with BMI 15 experienced a longer PFS (6.5 months) and OS (10.45 months) than patients with BMI < 15 (2.57 months and 5.63 months, respectively); conversely, PS ECOG status showed a significant association only with OS. The survival curves of in relation to BMI are shown in the figure 2. Table 4. Multivariable Cox regression analysis of PFS and OS for prognostic factors PFS OS HR (95%CI) for progression p value HR (95% CI) for mortality BMI < 15 HR 5.98 < 0.0001 5.38 (2.52-14.17) (2.02-14.31) 3 metastatic sites HR 2.48 < 0.0001 HR 2.94 (1.15 5.3) (1.09 7.95) PS ECOG 1 HR 0.96 0.92 HR 3.30 (1.27 8.57) p value < 0.0001 0.0277 < 0.0001

8 of 13 151 152 153 154 155 156 2.3 Dose administration and Safety analysis Fifty hundred and eighty-one doses of the combination of paclitaxel and ramucirumab and ten infusions of ramucirumab alone due to taxane-related toxicity were administered. The median number of paclitaxel and ramucirumab cycles was 4.12 (range 1.0 to 15). Disease

9 of 13 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 progression was the primary reason for treatment discontinuation. Ramucirumab-related toxicities were responsible for three treatment discontinuations: in one case the treatment was continued with paclitaxel alone for further 6 cycles, while in the other two cases the radiological assessment showed a disease progression, so paclitaxel was also discontinued at the same time. Details on toxicity profile are shown in table 5. Peripheral neuropathy related to paclitaxel was the most common adverse event and, as expected, it was associated with cumulative dose; due to grade 3 neuropathy, two patients required dose reduction of paclitaxel (60 mg/mq) and three patients needed to discontinue the taxane administration maintaining the treatment with ramucirumab alone. Other grade 3 non-hematological toxicities were uncommon and included one case of stomatitis and one case of hypertransaminasemia that were both responsible for treatment delay. The most common grade 3-4 hematological toxicity was neutropenia and it was the major cause of dose delay; however, no febrile neutropenia was observed and no patient required G-CFS support. Anemia was a common finding at the start of treatment and it was related to disease condition and previous chemotherapy treatments; two patients needed erythropoietin support but only one case of worsening to G3 toxicity (hemoglobin < 8.0 gr/dl) was observed requiring blood transfusion. One patient had G3 thrombocytopenia combined with G3 neutropenia. Among adverse events of interest related to the anti-angiogenic activity of ramucirumab, the incidence of any grade hypertension and bleeding was low. We had only one case of bleeding of the primary tumor requiring endoscopic argon plasma coagulation and one case of gastric perforation occurring five days after the last dose of ramucirumab in a woman who had required an implementation of corticosteroids due to brain metastases progression. One male patient experienced partial loss of vision caused by micro-embolization of the optical nerve; the event cleared up spontaneously after few days but led to treatment discontinuation. Table 5. Toxicity profile Event Grade 1 Grade 2 Grade 3 Grade 4 Hematological Anemia 6 4 1 0 Thrombocytopenia 1 0 1 0 Neutropenia 4 5 4 1 Non hematological Peripheral neuropathy 10 6 2 0 Stomatitis 1 1 0 0 Hypertransaminasemia 0 0 1 0 Diarrhea 2 2 0 0 Asthenia 5 2 1 0 Special interest (anti VEGFR activity) Hypertension 5 0 1 0 Bleeding 4 0 1 0 Gastrointestinal perforation 0 0 0 1 Embolism 0 1 1 0

10 of 13 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 3. Discussion Despite the increasing number of available agents in oncology, the clinical management of patients with mgc/mgojc is still disappointing and the prognosis remains poor [6]. After the results of RAINBOW trial, the combination of paclitaxel and ramucirumab has become the preferable therapy for patients who progress on platinum-based therapy. The aim of our analysis was to assess the activity and safety of this drug combination in our clinical daily practice. We are aware that the limited number of patients does not allow any definitive conclusion or generalization of the results; however, the validation of efficacy and safety outcomes in an unselected and heterogeneous patient population gives to this combination more sense and relevance in the real life practice. Despite the likely underestimation of overall survival due to the short time of median follow-up and the number of patients still on treatment, the median PFS (5.4 months) seems longer than that reported in the RAINBOW trial and in several previous reports. Taking into consideration the poor survival generally observed in Western population, the PFS in our study not only confirms the results achieved in the RAINBOW trial, but also suggests that they are reproducible in a general population with unfavorable prognosis. The clinical value of the combination of RAM and PXT resulted also in a recently published phase II study with nab-paclitaxel instead of paclitaxel carried out in an Asian less intensively pre-treated population with mainly gastric cancer, in which ORR and mpfs were even more higher (table 6) [7-9]. Table 6. Comparison with RAINBOW trial (RAM + PXT arm) and other single arm studies Outcomes RAINBOW Western pts RAMoss Bando et al Present paper Median OS (mo.) 9.6 8.6 8.3 n.r. 8.3 (*) Median PFS (mo) 4.4 4.2 4.5 7.6 5.4 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 ORR (%) 28 26.8 20.3 54.8 24.4 DCR (%) 80 76.8 59.7 92.9 61 (*) Median follow-up: 9.6 mo Subgroup analysis confirms the well-known positive prognostic factors affecting survival; however, the most relevant finding in our study is the strong association between PFS and BMI index. As a matter of fact, a better nutritional status implies a longer overall survival, but in our patients normal BMI was related not only to overall survival but also to PFS, an outcome that is primarily influenced by drug activity. The relevance of nutritional status as independent prognostic factor for PFS is confirmed by the lack of statistical relationship with performance status. Other reports have already focused the impact of malnutrition on chemotherapy dose intensity and toxicity incidence, but in our study no significant difference was observed in subgroup analysis [10, 11]. Therefore, we hypothesize a relationship between drug activity and nutritional status. One assumption may be that malnutrition could affect drug distribution and metabolism due to low levels of serum albumin and lower enzymatic activity. On the other hand, it is known that IgG monoclonal antibodies like ramucirumab are able to recall antibody-dependent cell-mediated cytotoxicity (ADCC) that might be impaired in malnourished patients [12]. Interestingly, the median duration of response is longer in our retrospective study than in the RAINBOW trial (5.4 months vs 4.4 months), despite the disease control rate is lower; this result may suggest that some patients obtain a greater benefit from the treatment. As previously underlined, our

11 of 13 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 HER-2 positive patients achieved a longer duration of response, according to similar data recently published in an Italian-Canadian paper [13]. Although the cohort was very small (12 patients) for definitive conclusions and the huge number of ongoing treatments likely affected outcomes estimations, the Cox analysis showed a trend toward an association with a longer PFS (6.8 vs 5.27 months; HR 0.65, p = 0.1341.). These data suggest a possible relationship between HER-2 overexpression and downstream activation of the VEGF pathway within gastric tumor cells; an overlap of these pathways has been demonstrated in in-vitro models of HER-2 breast cancer [14-16]. We did not find that the presence of peritoneal metastases is a negative prognostic factor; since some patients experienced disease progression due only to the worsening of peritoneal involvement and ascites, we think that treatment of peritoneal disease by locoregional procedures (HIPEC, PIPAC) could be a useful option in the management of selected cases. The combination of paclitaxel and ramucirumab showed a favorable safety profile even in this unselected and heavily pre-treated population of patients. Grade 3-4 AEs leading to dose reduction or withdrawal were relatively rare. Only two severe adverse events may be related to the class of anti-angiogenic agents, including one case of gastric perforation following the increase of corticosteroid medication due to brain metastases. Low incidence of these AEs is noteworthy particularly considering that half of patients do not have undergone gastric surgery. 4. Materials and Methods We retrospectively collected clinical and radiological data of patients with metastatic gastric and gastro-oesophageal junction adenocarcinoma treated with paclitaxel and ramucirumab as second or third line therapy at Medical Oncology Department of Policlinico Universitario A. Gemelli in Rome. Medical records were reviewed in order to obtain information about demography, treatment, outcome and safety. CT scan were performed about every 12 weeks or less in cases in which there was evidence of clinical deterioration. Between November 2015 and March 2018, 47 patients were treated with ramucirumab (8 mg/kg intravenously on days 1 and 15) plus paclitaxel (80 mg/m² intravenously on days 1, 8, and 15 of a 28-day cycle) for metastatic GOJC/GC after failure of previous platinum-based therapy. Drugs were mostly infused in outpatient department and the treatment was continued until disease progression or unacceptable toxicity. Prophylactic antiemetic therapy with steroids and 5-HT3 receptor antagonists was administered before each cycle according to routine clinical practice. Primary prophylaxis with C-CSF was not planned. Disease responses were classified according to RECIST criteria (version 1.1). Overall survival was defined as the time interval from the start of therapy to the date of death or last follow-up visit. Progression-free survival was defined as the time interval from the first day of therapy to the date of clinical or radiological disease progression or death, whichever occurred first. OS and PFS were estimated using the Kaplan-Meier method. Prognostic factors were analyzed using Cox proportional hazards regression models in 2 steps. Firstly, each potential prognostic factor was screened as indipendent covariate. Then, the final prognostic factors were identified based on a multivariable Cox model that was built using stepwise selection of covariates (entry significance level 0.05, exit significance level = > 0.1). All data were analyzed using statistical software SPSS 20 for Windows (IBM Corp., NY, USA). Toxicities were reported according to the CTCAE criteria version 4.0. 5. Conclusions In conclusion, our study confirms the efficacy and safety of the combination of ramucirumab with paclitaxel also in real-life practice. These findings need further validation in a larger cohort of patients with a longer follow-up; we think that the significant impact of nutrition on survival deserves a particular attention especially if it would be related to drug activity rather than to patients frailty. It implies that nutritional counselling should be part of clinical practice for patients with gastric cancer and that an accurate assessment of nutritional parameters, including also sarcopenia and lack of nutrients, would be necessary in planning future clinical studies. Finally, the possible

12 of 13 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 relationship of HER-2 expression to PFS raises the important question of identifying predictive biomarkers of response to ramucirumab, including HER-2 status and exposure to trastuzumab [17,18]. As result of expanding therapeutic options, our report underlines also the importance both of the oncologist experience and of the multidisciplinary approach in clinical decision-making process for patients with gastric cancer. 6. Compliance with ethical standards Conflict of interest. No funding sources were used in the preparation of this manuscript and the authors declare that they have no conflicts of interest. Author Contributions. First two authors contributed equally to this manuscript Conceptualization, Antonia Strippoli and Alessandra Cassano; Data curation, Emanuele Vita, Antonia Strippoli, Vincenzo Di Noia, Carmela Di Dio, Valeria Zurlo, Michele Basso, Ettore D'Argento and Giovanni Schinzari; Formal analysis, Emanuele Vita and Vincenzo Di Noia; Investigation, Emanuele Vita, Antonia Strippoli, Vincenzo Di Noia, Carmela Di Dio, Valeria Zurlo, Michele Basso, Ettore D'Argento and Giovanni Schinzari; Methodology, Emanuele Vita and Vincenzo Di Noia; Project administration, Antonia Strippoli and Alessandra Cassano; Supervision, Michele Basso, Ettore D'Argento, Giovanni Schinzari, Carlo Barone and Alessandra Cassano; Validation, Carlo Barone and Alessandra Cassano; Writing original draft, Emanuele Vita and Antonia Strippoli; Writing review & editing, Emanuele Vita, Antonia Strippoli, Carlo Barone and Alessandra Cassano. Ethical approval. No ethical approval or informed consent statement was required for this review article. 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 References 1. Siegel RL, Miller KD, Cancer statistics, 2017. CA Cancer J Clin 2017. 67(1):7-30. doi: 10.3322/caac.21387. 2. AIRTUM AIOM. I numeri del cancro in Italia 2017 3. Wagner AD, Grothe W, Chemotherapy in advanced gastric cancer: A systematic review and meta-analysis based on aggregate data. J Clin Oncol 2006. 24(18):2903-9 doi: 10.1200/JCO.2005.05.0245 4. Wilke H, Muro K, Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised Phase 3 trial. Lancet Oncol 2014. 15(11):1224-35 doi: 10.1016/S1470-2045(14)70420-6 5. Fuchs CS, Tomasek J, Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, Phase 3 trial. Lancet 2014. 383(9911):31-39 doi: 10.1016/S0140-6736(13)61719-5 6. Van Cutsem E, Van de Velde C, Expert opinion on management of gastric and gastrooesophageal junction adenocarcinoma on behalf of the European Organisation for Research and Treatment of Cancer (EORTC)-gastrointestinal cancer group. Eur J Cancer. 2008. 44(2):182-94 doi: 10.1016/j.ejca.2007.11.001

13 of 13 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 7. Shitara K, Muro K, Subgroup analyses of the safety and efficacy of ramucirumab in Japanese and Western patients in RAINBOW: a randomized clinical trial in second-line treatment of gastric cancer. Gastric Cancer. 2016. 19(3):927-38. doi: 10.1007/s10120-015-0559-z 8. M. Di Bartolomeo, M. Niger, Safety and efficacy profile of ramucirumab alone or combined with paclitaxel in metastatic gastric cancer (MGC): a real-life overview of compassionate-use named patients (pts) (RAMoss study). Ann Oncol 2016. 13(2):227-234. doi: 10.1093/annonc/mdw371 9. Bando H, Shimodaira H, A phase II study of nab-paclitaxel in combination with ramucirumab in patients with previously treated advanced gastric cancer. Eur J Cancer 2018, 91:86-91. doi: 10.1016/j.ejca.2017.11.032 10. Klute KA, Brouwer J, Chemotherapy dose intensity predicted by baseline nutrition assessment in gastrointestinal malignancies: a multicentre analysis. Eur J Cancer 2016, 63:189-200. doi: 10.1016/j.ejca.2016.05.011 11. Qiu M, Zhou YX, Jin Y, Nutrition support can bring survival benefit to high nutrition risk gastric cancer patients who received chemotherapy. Support Care Cancer 2015, 23(7):1933-9. doi: 10.1007/s00520-014-2523-6 12. Vacchelli E, Pol J, Trial watch: Tumor-targeting monoclonal antibodies for oncological indications. Oncoimmunology 2015, 4(1):e985940. doi: 10.4161/2162402X.2014.98594 13. Tehfe M, Tabchi S, Ramucirumab in HER-2-positive gastroesophageal adenocarcinoma: an argument for overcoming trastuzumab resistance. Future Oncol. 2018, 14(3):223-228 doi: 10.2217/fon-2017-0434 14. Xiong S, Grijalva R, Zhang L, Nguyen NT, Pisters PW, Pollock RE et al. Up-regulation of vascular endothelial growth factor in breast cancer cells by the heregulin-β1-activated p38 signaling pathway enhances endothelial cell migration. Cancer Res. 2001, 61(4), 1727 1732 15. Tsai PW, Shiah SG, Up-regulation of vascular endothelial growth factor C in breast cancer cells by heregulin-beta 1. A critical role of p38/nuclear factor-kappa B signaling pathway. J Biol Chem. 2003, 278(8):5750-9. doi: 10.1074/jbc.M204863200 16. Klos KS, Wyszomierski, ErbB2 increases vascular endothelial growth factor protein synthesis via activation of mammalian target of rapamycin/p70s6k leading to increased angiogenesis and spontaneous metastasis of human breast cancer cells. Cancer Res. 2006, 66(4):2028-37. doi: 10.1158/0008-5472.CAN-04-4559 17. Fuchs CS, Tabernero J, Biomarker analyses in REGARD gastric/gej carcinoma patients treated with VEGFR2-targeted antibody ramucirumab. Br. J. Cancer 2016, 115(8):974-982. doi: 10.1038/bjc.2016.293 18. Fuchs CS, Muro K, Prognostic Factor Analysis of Overall Survival in Gastric Cancer from Two Phase III Studies of Second-line Ramucirumab (REGARD and RAINBOW) Using Pooled Patient Data. J Gastric Cancer. 2017, 17(2):132-144 doi: 10.5230/jgc.2017.17.e16.