Next Article in Journal
Real-World Study to Assess Patterns of Treatment Practices and Clinical Outcomes in Metastatic Colorectal Cancer Patients with RAS Wild-Type Left-Sided Tumours in Canada
Next Article in Special Issue
In-Clinic versus Hybrid Cancer Rehabilitation Service Delivery during the COVID-19 Pandemic: An Outcome Comparison Study
Previous Article in Journal
Presence and Role of Associations of Cancer Patients and Volunteers in Specialist Breast Centres: An Italian National Survey of Breast Centres Associated with Senonetwork
Previous Article in Special Issue
Cancer Survivors Living in Rural Settings: A Qualitative Exploration of Concerns, Positive Experiences and Suggestions for Improvements in Survivorship Care
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Mistletoe Extracts during the Oncological Perioperative Period: A Systematic Review and Meta-Analysis of Human Randomized Controlled Trials

1
Patterson Institute for Integrative Oncology Research, Canadian College of Naturopathic Medicine, Toronto, ON M2K 1E2, Canada
2
Radiation Oncology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
3
The Centre for Health Innovation, Ottawa, ON K2P 0M7, Canada
4
Pacific College of Health Sciences, San Diego, CA 92108, USA
5
National Centre for Naturopathic Medicine, Southern Cross University, Lismore 2480, Australia
6
School of Public Health, University of Technology Sydney, Ultimo 2007, Australia
7
The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2023, 30(9), 8196-8219; https://doi.org/10.3390/curroncol30090595
Submission received: 18 July 2023 / Revised: 11 August 2023 / Accepted: 22 August 2023 / Published: 6 September 2023
(This article belongs to the Special Issue Optimizing Integrated Cancer Care from Diagnosis to Survivorship)

Abstract

:
Background: We aim to evaluate the safety and efficacy of mistletoe extract (ME) use during the oncological perioperative period. Methods: Details registered a priori on PROSPERO (CRD42018086168). Results: Seven RCTs (comprising 663 participants in nine reports) and three nonrandomized studies were included. In five RCTs, ME was evaluated as adjunctive care and the control group had no additional intervention, whereas in two RCTs, ME was compared head-to-head against common cancer treatments (i.e., etoposide or bacillus Calmette-Guérin) with the intervention groups not receiving standard care. Meta-analyses found no evidence for a difference between ME and no added therapy for mortality and recurrence (RR, 95% CI: 1.00, 0.79–1.27; and 1.03, 0.79–1.33, respectively). Two RCTs reported beneficial effects of ME on immune cells, specifically natural killer cells, in colorectal cancer, and one RCT reported quality of life improvement. Two RCTs reported ME discontinuations due to adverse events and grade 3/4 toxicities. Nevertheless, no safety signals were detected from these 10 studies. Quality appraisal revealed a substantial risk of bias. Conclusions: Preliminary data are encouraging for mistletoe extracts, particularly in the context of colorectal cancer. However, the evidence is limited by the number of studies, an evaluation of different outcomes, and methodological limitations. Further high-quality research is warranted.

1. Introduction

Mistletoe is a parasitic plant that grows on trees without harming the host [1,2,3,4], and it has been widely used as part of traditional medicine in Europe and in other parts of the world to treat several health conditions [4,5,6]. In contemporary settings, the most common routes of administration are via subcutaneous (SC) or intravenous (IV) injection. Other routes of administration have also been reported [1,3].
The application of mistletoe was first proposed for use in cancer by Rudolf Steiner in 1920 [7]. Following this, the clinical use of mistletoe extract (ME) in cancer as an adjuvant tool has been further explored and expanded [8]. Viscum album L. extracts (VAEs, V. album L.; European mistletoe) are the most commonly used species due to their potential anti-cancer properties demonstrated in vitro [2,9].
There are many bioactive compounds in ME that have been characterized, including various lectins (I, II, III), viscotoxins, amino acids, flavonoids and polysaccharides [10]. Among these compounds, those believed to be most relevant to cancer-supportive care are mistletoe lectins and viscotoxins [11]. Viscotoxins are small proteins that mainly have cytotoxic (cell-killing) effects and possible immune system stimulatory properties [12], whereas Lectins are complex molecules consisting of both protein and carbohydrates and can change the biochemical characteristics of immune cells and modulate their functions by binding to their outside surface [13,14]. Mistletoe extract preparations have demonstrated both cytotoxic and immune-stimulatory effects against tumor cells in vitro [15,16,17,18].
The cytotoxic effect of ME and its constituents may occur by targeting two signaling pathways that regulate tumor proliferation resulting in apoptosis and cancer cell cycling, namely the PI3K/AKT (phosphatidylinositol 3-kinase/protein kinase B) and MAPK (mitogen-activated protein kinase) pathways [4,19,20,21,22]. Other mechanisms by which mistletoe may exert its cytotoxic effect are through the promotion of apoptosis [21], arresting cell cycling [22], inhibiting angiogenesis [23], generation of reactive oxygen intermediates (ROI) and inhibiting cellular protein synthesis [24,25,26].
Dendritic cells (DCs) and natural killer (NK) cells are the primary cell subtypes of the immune system that appear to be most positively affected by mistletoe. The use of Viscum album extracts (VAEs) on immature dendritic cells led to stimulation of both the maturation and activity of DCs in one study [27]. Similarly, Korean mistletoe lectins (Viscum album coloratum) have enhanced the maturation of bone-marrow-derived dendritic cells in vitro [28]. Tumor cells have been able to resist immune-mediated destruction by producing immunosuppressive substances that can reduce dendritic cell activity. In addition to enhancing dendritic cell maturation, mistletoe may counteract DC immunosuppression, as evidenced by a renal cancer cell model [29].
Another effect on the immune system demonstrated by MEs, particularly lectin-rich extracts, is its action on NK cells. This has been demonstrated through enhanced lysis of glioblastoma cells in an in vitro study [30], and in another study, an increase in natural killer cytotoxic activity among breast cancer patients after a single intravenous infusion of a Viscum album extract [31]. In cultures of human peripheral blood mononuclear cells exposed to ME the secretion of immune-system-enhancing cytokines, such as interleukin-1, interleukin-6, and tumor necrosis factor-alpha were increased [16,32].
Despite the benefits of surgery, it can have a profound impact on a person’s physiology and immune function and can negatively affect the course of cancer, including its recurrence. Surgery and cancer often lead to immunosuppression and, together, they can have an additive immunosuppressive effect and consequently lead to an increased risk of postoperative complications such as infections and potentially the spread of malignancy [15]. Immunomodulation (and the use of immunotherapy) in the perioperative period before, during or after surgery may have an important role in overcoming these risks [15].
The inflammatory response to surgery or trauma creates surgical inflammation [33,34,35]. The local inflammatory response in a surgical wound occurs in the early postoperative period and leads to the activation of the innate immune response. Consequently, there is an influx of neutrophils and monocytes into the wound and a major production of cytokines and chemokines [36]. In the postoperative period, a balance is required between pro-inflammatory cytokines and anti-inflammatory agents and cytokine release. This balance helps to restore and maintain hemostasis and prevent systemic inflammatory immune response (SIRS) or immunosuppression, respectively [36,37,38,39]. Mistletoe, through inhibition of the COX protein, may act as an anti-inflammatory agent in this setting [40].
Surgery could influence local recurrence and spread of cancer cells as shown from in vivo experimental research where the increased severity of surgical stress enhances the development and severity of the lung metastases [41]. Furthermore, animal studies suggest that stress hormones released in response to surgery could be a risk factor for cancer recurrence [42]. In addition, surgery promotes a hypercoagulable state [43,44]. This hypercoagulable state decreases activity of NK cells and increases the survival of tumor cells or decreases the clearance of the tumor cell, thus promoting metastases [44]. Moreover, low pre-operative NK cell activity was found to be associated with tumor recurrence in colorectal patients who underwent surgery [45].
Cell-mediated immunosuppression during the perioperative period could also help promote the development of metastases after surgery [46,47]. This suppression of cellular immunity following major surgery is transient, yet can be clinically significant [47]. In colorectal cancer patients who underwent surgery, NK cell activity dropped by day one postoperatively and only started to recover in the third week post-surgery; this correlated with the burden of the disease [45]. As a result, the early postoperative period is suggested to be an ideal time for immune-based interventions that reduce or protect against immunosuppression [46].
Of added interest in this area is that tumors produce anti-angiogenic as well as local pro-angiogenic activators [48]. Upon surgery, there may be a reduction in the production of more stable anti-angiogenic mediators like angiostatin or endostatin, as well as post-surgical production of angiogenesis signals, leading to potential angiogenesis of previously dormant micrometastases [49,50]. In this context, the use of anti-angiogenic therapies may in fact have a role to play in mitigating some of the risks of surgery [51].
Surveys suggest that mistletoe therapy remains a relatively popular choice for cancer patients [52,53]. However, despite increasing consumer familiarity and adoption of ME alongside other integrative medicine therapies in cancer care [54], ME has not been approved for use in cancer care in some jurisdictions [16]. Prior studies and related systematic reviews offer mixed or inconclusive conclusions regarding the effectiveness of ME, perhaps due to variations in preparations and their pharmacological properties. In addition, the inadequate methodological design of many studies creates more uncertainty as to the clinical utility of this therapy [55,56].
To address this gap, we aim to evaluate whether ME use in cancer patients around surgery may help overcome the undesired effects of surgery mentioned earlier and to identify any potential effects on the cancer itself. We have conducted this systematic review and meta-analysis to synthesize available evidence, determine the potential role of this therapy in the perioperative setting, and determine what additional research is needed. To our knowledge, this is the first systematic review and meta-analysis that evaluates the efficacy and safety of the use of ME in cancer patients during the perioperative period.

2. Materials and Methods

This systematic review is part of a larger umbrella project to prepare systematic reviews examining the use of natural health products among cancer patients during the perioperative period and was based on our institution’s initial prioritization exercise [57]. The current report describes the mistletoe section of this larger research agenda. The original protocol was registered a priori on the PROSPERO website (CRD42018086168) and has been described in two previous publications in detail [58,59]. The conduct of this systematic review followed the Cochrane Collaboration guidelines [60] and was written following PRISMA reporting guidance [61,62].
The inclusion criteria for this systematic review allowed for patients undergoing cancer-related surgery that evaluated mistletoe extracts, utilizing any method of administration, formulation, duration, and dose compared to placebo, standard care, or active control. The primary outcomes assessed were cancer recurrence, mortality, treatment response, remission, stable disease, and metastasis/disease progression. Secondary outcomes included postoperative complications, postoperative infections, adverse events, blood loss (perioperative blood transfusion), length of hospitalization, pain, fatigue, quality of life (performance status), anthropometric indices, immune cell measurements, inflammatory markers levels, and cancer biomarkers. We included published and unpublished reports concerning the use of mistletoe in cancer patients during the perioperative period.
To assess for efficacy outcomes, only randomized controlled trials (RCTs) were included, whereas for safety outcome evaluation, non-randomized controlled clinical trials and controlled observational studies, in addition to the RCTs, were included for broader data capture. Non-English reports and studies examining the use of concomitant conventional treatment (i.e., chemotherapy, radiotherapy, etc.) with mistletoe were excluded.
MEDLINE, Embase, and Cochrane CENTRAL databases were searched for human studies in English from inception to 14 January 2023. The Peer Review of Electronic Search Strategies (PRESS) checklist [63] was used by an expert librarian (JM) to peer review the literature search (the Supplementary File includes the MEDLINE search strategy). Furthermore, searches were performed within the clinicaltrials.gov (accessed on 17 July 2023) trials registry, the Natural Medicine (formerly Natural Standard) database, and Health Canada’s Natural Health Product Monographs as a supplemental grey literature search. To identify any other potentially relevant studies, we scanned the list of references of the included studies and the related systematic reviews.
Screening studies for eligibility, data extraction, and risk of bias appraisal of the RCTs and quality assessment of the non-RCT and observational studies were conducted independently by two research team members with a consensus approach taken to resolve any discrepancies. The original (2011) Cochrane Risk of Bias tool was used for appraisal of the RCTs [64]. For the assessment of quality for non-RCT and observational studies, the Newcastle–Ottawa Scale was applied (NOS) [65].
Data extraction was performed in Excel sheets; descriptive synthesis of the result was conducted first and then evidence summary tables were created. Meta-analyses were conducted when possible and appropriate using STATA 12 [66]. The guidelines from the Council for International Organizations of Medical Sciences (CIOMS Working Group X) and Cochrane were followed in dealing with statistical heterogeneity and in conducting the meta-analysis [60,67]. Forest plots were created to graphically present the main results from the meta-analysis. The relative risk (RR) for binary outcomes and mean difference (MD) for continuous outcomes were calculated for each trial, when possible, to represent the summary effect measures.

3. Results

3.1. Included Studies

The details of the literature search, including title and abstract screening, full-article screening, reasons for exclusion within the larger project, and the number and type of studies for the mistletoe analysis, are presented in the study flow diagram in Figure 1. In summary, the searches retrieved 4952 records (abstracts) for the larger project, and 442 full-text articles were screened. Overall, 290 studies were eligible and included. For the mistletoe section, twelve relevant articles were included reporting on ten studies comprising seven RCTs in nine reports [15,68,69,70,71,72,73,74,75] and three observational cohort studies [76,77,78]. The seven RCTs included 633 participants in total.
The trial and patient characteristics of the included RCTs and the non-RCTs are reported in Table 1 and Table S1, respectively. Additionally, details of the intervention and control arms of the RCTs and non-RCTs are outlined, respectively, in Table 2 and Table S2. Summary characteristics across all studies are presented in Table S3. The RCTs’ publication dates ranged from 2001 to 2020, with all seven RCTs initially published between 2001 and 2009. Two additional updates from one RCT [71] were published in 2014 and 2020 [72,73]. Sample sizes of the RCTs ranged between 20 and 204 participants. Three RCTs (43%) had a sample size of less than 50 patients. Six RCTs were conducted in Europe, and one was conducted in Egypt. Funding was reported in six RCTs; three (43%) were publicly funded, one RCT (14%) was privately funded, one RCT (14%) had a mix of public and private funding, and one RCT (14%) did not have a funding source. In one RCT (14%), funding was not reported.
The cancer populations studied in the seven RCTs included two (29%) for bladder [68,69], two for digestive tract/colorectal cancer [15,74] and one each (14%) for osteosarcoma [73], melanoma [70] and head and neck cancer [75]. The mean or the median age of the study populations among six RCTs ranged between 33.9 and 71 years old. One RCT did not report the mean or median age of the sample’s population for each arm, but rather reported that the mean age in the ME group was 62 years (range 37–87 years) with no differences between the groups regarding the age. The proportion of females among six RCTs ranged between 9% and 45%, and no data regarding the percentage of the female population were reported in one RCT.
ME was administered during the postoperative period in four RCTs (57%), during both pre-and postoperative periods in two RCTs (29%), and intra-operatively in one RCT (14%). Duration of administration in the RCTs ranged from a single intravenous application intra-operatively (administered over a one-hour time period) [74] up to 60 weeks (15 months, given in cycles with 12 weeks of treatment followed by 4 weeks without treatment) [75].
The majority of RCTs (57%) compared mistletoe to usual care (i.e., with no added treatment) [15,68,70,74,75]. However, in two RCTs (29%), mistletoe was not evaluated as adjunctive but instead compared against a common cancer treatment (i.e., etoposide or bacillus Calmette-Guérin [BCG]), so the intervention group did not also receive standard care [69,73]. The subcutaneous (SC) administration of mistletoe was the most common route of administration with five RCTs (72%) utilizing this application method. One RCT (14%) used mistletoe intravenously (IV) intra-operatively, and one RCT reported intravesical application in bladder cancer patients.

3.2. Methodological Quality Assessment and Risk of Bias

Table 3 and Figure 2 depict the risk of bias assessments for each RCT and the aggregate summary across the RCTs, respectively, using the Cochrane Risk of Bias tool. A substantial risk of bias was detected among all the included studies using the methodological quality appraisals as none of the RCTS were judged to have a low risk of bias overall. Four (57%) RCTs were rated as high risk of bias for at least one domain among the seven assessment elements. The other three (43%) RCTs had an unclear risk of bias for at least one domain. The elements that scored worst were related to blinding as none of the RCTs reported being double-blinded, suggesting potential performance and detection bias. In addition, the majority of the studies were unclear for selective reporting as trial registration (or an accessible protocol) was not cited, indicating possible reporting bias.
Table 4 presents the quality assessment of the three non-RCTs using the Newcastle–Ottawa Scale (NOS). Methodological quality varied considerably, with the Augustin study [77] scoring best with nine out of nine total stars. However, the “Outcome” domain demonstrated several limitations in the Bussing and Elsasser-Beile studies [76,78], in particular due to short follow-up times. Moreover, the “comparability of cohorts on the basis of the design or analysis” item was inadequate as these two studies did not control for important potential confounding. Regarding publication bias, while there were too few studies to reliably create a funnel plot, this concept is incorporated in the original Cochrane ROB tool (i.e., non-reporting, missing results).

3.3. Results of the RCTs by Comparators

The outcomes of the RCTs are presented according to the control group and divided into these three relevant comparison categories:
  • Mistletoe versus no added treatments;
  • Mistletoe versus etoposide;
  • Mistletoe versus BCG.

3.3.1. Mistletoe Versus No Added Treatments (5 RCTs)

Mistletoe was compared to no added treatment in five out of seven RCTs (72%) [15,68,70,74,75].

Primary Outcomes (First Comparison)

  • Mortality, Recurrence and Metastasis
Three RCTs in this comparison evaluated mortality and were included in the meta-analysis [68,70,75]. Supplementary Table S4 presents the mortality outcome data and Figure 3 displays the forest plot of the meta-analysis for mortality. A fixed-effects model was applied as there was low statistical heterogeneity between studies (I2 = 0%). The follow-up duration among the studies ranged between 18 months and 8 years, with one study noting a median follow-up period of 4 years [75]. Meta-analysis found no evidence of a difference in survival between ME and no added treatment (number of studies, k = 3; total sample size, N = 451; RR = 1.00; 95% CI, 0.79 to 1.27; p = 0.97). In addition, over the Kleeberg 2004 RCT [70] 8-year follow-up period in melanoma patients, there were slightly more deaths in the ME group (59% vs. 53% of participants), but the difference was not statistically significant, and the total number of deaths among the three RCTs was very similar (74 vs. 75, respectively).
Figure 4 presents the forest plot of the meta-analysis for cancer recurrence. Similar to the mortality outcome for this comparison, three studies reported on cancer recurrence (the same studies reported on mortality outcome) and were included in the meta-analysis [68,70,75]. Follow-up time for assessing cancer recurrence varied between 18 months to 8 years. A random effects model was applied for meta-analysis as there was moderate statistical heterogeneity between the studies (I2 = 36%). The comparison of mistletoe versus no added treatment yielded no evidence of a difference in cancer recurrence (k = 3; N = 451; RR = 1.03; 95% CI, 0.79 to 1.33; p = 0.85). Metastasis outcome data were included in the recurrence meta-analysis data mentioned above, which revealed no evidence of a difference between the mistletoe and control group for recurrence (including metastasis). Two RCTs in this comparison reported on the incidence of distant metastasis. No other extractable data were reported within the five RCTs for this comparison regarding the other primary outcomes of the systematic review including treatment response, remission, and stable disease.

Secondary Outcomes (First Comparison)

  • Quality of Life
One RCT reported on quality of life for this comparison at 60 days follow-up. Enesel 2005 found that ME (Isorel) administration (n = 40) improved self-reported functional impairment score (measured with the Karnofsky Performance Index (KPI)) compared to baseline value by 22% (p < 0.01) in participants with digestive tract cancers (51% colorectal), while the KPI decreased by 13% (p < 0.05) in the control group (n = 30) [15]. Quality of life results, including for fatigue and pain subscales, reported for all RCTs are presented in Table S4.
  • Immune Cells and Inflammatory Markers
Two RCTs investigated the effects of ME on immune cells and inflammatory markers for this comparison. The results of the immune cells and inflammatory markers for all RCTs are presented in Table S4. Enesel 2005 evaluated over 15 blood parameters in digestive tract cancer participants at 28-day follow-up (i.e., 14 days postoperative). Compared to baseline values (i.e., those reported as within-group relative percent change), the ME arm had increased numbers of NK cells (by 44%, determination method using CD2-CD3) and T and B cells. In particular, the increases were seen in T-helper (CD4) cell (by 18%), complement (by 20–30%), IgA (by 27%), and IgM (by 23%) values after Isorel injections for 2 weeks pre-operatively and 2 weeks postoperatively. In contrast, the no added treatment control group had a non-significant decrease in NK values and had a greater increase in total peripheral leukocyte (WBC) count compared to their baseline mean values. Notably, CD8 (T-suppressor) values in the ME group showed a significant decrease (by −7%) compared to baseline, whereas in the control arm, there was a slight but non-significant increase (by 3%) [15]. Schink (2007) examined intra-operative ME use in colorectal cancer and found a smaller drop from baseline in NK cell activity on day 7 postoperatively in the mistletoe group versus the control (p = 0.04). In regard to monocyte expression of HLA-DR antigen in this RCT, there was no difference seen between the mistletoe and control groups (p = 0.46) [74].
  • Blood Loss
One study reported on perioperative blood transfusion (erythrocyte administration) in this comparison (Table S4) and found no significant difference in the number of perioperative blood transfusion events: 1 in 12 patients in the ME (Iscador® M) group versus 3 in 14 patients in the no added treatment group (p > 0.05) [74].
  • Other Complications and Adverse Events
For this comparison, four RCTs [68,70,74,75] reported on adverse events (AEs) and other complications while one RCT did not report on adverse events [15]. Table 5 presents detailed adverse event data reported across all RCTs. Treatment discontinuations due to grade 3 or 4 (WHO) toxicities in one RCT among melanoma patients were more frequent in the ME arm versus no added treatment (5 out of 102 vs. 0 out of 102 participants, respectively) [70]. One RCT reported no adverse events after application of mistletoe [68]. Another RCT found similar numbers between groups for AEs during surgery, serious AEs, and postoperative AEs [74]. The final RCT did not report any AEs in the control arm, whereas in the ME group, 16 out of 97 head and neck cancer participants refused further injections because of mistletoe-induced AEs. Local and/or systemic reactions around the time of injection were reported in 47 out of 97 (with rubor and prurigo being the most common) at the start of therapy and at 32 weeks or later in 4/97 patients. Among the same ME cohort, general drug reactions occurred in 4 out of 97 patients (e.g., melalgia, fever ≤ 39 °C, sleeplessness, tiredness, coldness or heat sensation, sneezing) [75]. No extractable data were reported in these five RCTs regarding the other secondary outcomes of the review, namely postoperative infection, hospitalization length of stay, anthropometric data (BMI and body weight), cancer biomarkers, fatigue, and pain.

3.3.2. Mistletoe versus Etoposide (Second Comparison (1 RCT)

Primary Outcomes (Second Comparison)

The Longhi et al. RCT, which is reported across three publications, includes a report on a 12-month trial follow-up period among a small sample of patients with osteosarcoma randomized to either ME (no other conventional treatment given) or etoposide after surgery for a second cancer relapse in the lung. The number of deaths due to any cause was 0 out of 9 patients in the mistletoe group versus 3 out of the 11 patients (27%) in the etoposide group (p > 0.05). In addition, the number of patients with a relapse after one year was 4 out of 9 (44%) in the mistletoe group versus 8 patients out of 11 (73%) in the etoposide group (p > 0.05) [71,72,73].

Secondary Outcomes (Second Comparison)

  • Quality of Life
Quality of life was assessed in the Longhi (2014) RCT using the EORTCBB QLQ-C30 questionnaire in adults or the Pediatric Quality of Life Cancer Module Acute version 3.0 (PedsQL) in patients less than 18 years old. There was a trend for positive effects from using ME on most of the parameters and in particular for the global QoL scale, functional scales including the physical and social functioning, and financial functioning, in addition to the symptom items of fatigue, pain, and dyspnea. In contrast, with the use of etoposide, improvement was only seen for social functioning and there were deteriorations in the pain and nausea/vomiting symptom items (Table S4) [72].
  • Immune Cells and Inflammatory Markers
Longhi (2020) reported in a small RCT that administered ME for 12 months versus etoposide for 6 months and compared lymphocyte subpopulation counts for CD3, CD4, and CD56 (NK) cells compared to baseline. In the ME arm, these all increased at 6 and 9 months and then decreased at 12-month follow-up (e.g., for CD56, mean and SD: –24.59 ± 180.06 mm3). Notably, only by the last timepoint had these increased in the standard drug group (N = 9; Table S4) [73].
  • Other Complications and Adverse Events (RCTs)
The etoposide drug group experienced more total adverse events and treatment discontinuations than the mistletoe extract arm in this RCT, but serious AEs (i.e., hospitalizations due to surgery) were similar. Details of the AEs and other complications are presented in Table 5. The following secondary outcomes of the review—postoperative infection, hospitalization length of stay, anthropometric data (BMI and body weight), blood loss, and cancer biomarkers—were not reported in this RCT.

3.3.3. Mistletoe versus BCG (1 RCT) (Third Comparison)

Primary Outcomes (Third Comparison)

  • Recurrences and Metastasis
Ibrahiem (2009) only reported results in a conference abstract format and reported recurrence rates after using intravesical mistletoe versus BCG (route not reported) in superficial bladder cancer patients (Ta, T1 of Grade 1–2) following TURBT (transurethral resection of bladder tumor). Five patients in each group developed invasive bladder cancer (p > 0.05). The number of local recurrences was 9 out of 30 patients in the BCG group versus 22 out of 30 patients in the mistletoe group (p = 0.003). Stage and grade progression occurred in 14 patients (47%) in the mistletoe group versus 4 patients (13%) in the BCG group (p = 0.01). One patient in the mistletoe group developed lymphatic and hepatic metastases versus no patients in the BCG group (p > 0.05) [69]. This RCT did not report on the rest of the systematic review’s primary outcomes.

Secondary Outcomes (Third Comparison)

  • Other Complications and Adverse Events (third comparison)
Self-limited local skin reaction after first SC dose was reported in all patients with ME, and the authors only reported that the intravesical ME was “tolerable and safe” without providing further details. A painful bladder sensation was reported in 23 of the 30 patients in the BCG group. Further details of the AEs and other complications are presented in Table 5. This RCT did not report on the other secondary review outcomes of interest.

3.4. Results of Non-Randomized and Observational Studies

Three non-RCTs were additionally included for further safety evaluation, comprising two non-randomized clinical trials and one observational controlled cohort study in participants with malignant melanoma (N = 686), breast cancer (N = 101), and superficial bladder cancer (N = 48) (Tables S1–S3). The primary outcomes of mortality, recurrence, and metastasis were only reported in one study each, and no safety concerns were seen (data presented in Supplementary Table S5). The only secondary review outcome of interest reported across these three studies, other than adverse events (AEs), was immune and inflammatory markers in one study, which found that C-reactive protein, leukocytes, lymphocytes and the main subsets (i.e., T cells, B cells, natural killer cells, CD4+, CD8+ CD28 and CD8+ CD28+ CD25+ T cells, and the cytokines interleukin 6) were similar between the ME and no added treatments groups [78]. Table S5 presents the AE data reported in these studies. Two non-randomized clinical trials reported that there were no mistletoe-related side effects among 80 total participants in the ME groups [76,78]. Augustin (2005) conducted a controlled cohort study evaluating 329 participants who had received mistletoe extract for a median of 30 months and reported 11 (3%) systemic adverse drug reactions (ADRs) attributed to the mistletoe treatment, 42 local injection site reactions, and 6 mistletoe treatment terminations due to ADRs [77].

4. Discussion

This comprehensive systematic review included 12 publications, in which seven RCTs with 663 participants were synthesized to evaluate ME efficacy in the context of perioperative cancer care. The results of this systematic review with meta-analysis suggest a possible benefit of ME use for patients with cancer undergoing surgery, with a positive impact on immune function (NK cells) and also on quality of life (functional impairment). This is primarily from data from two RCTs comparing ME against no added therapy. The evidence, however, is fairly sparse, and the risk of bias is unclear or high, mainly due to a lack of blinding and/or placebo/sham controls. Methodological limitations weaken the strength of the findings and further investigation is warranted using study designs that address these limitations.
The seven included RCTs were published between 2001 and 2009, and two additional updates of an original RCT were published in 2014 and 2020. Additionally, three non-randomized controlled studies were included on the safety of using ME in this context. There was clinical and methodological heterogeneity across the RCTs. The cancer types studied in the RCTs included the following: bladder (two), digestive tract/colorectal (two), osteosarcoma (one), melanoma (one), and head and neck cancer (one). Various extracts of mistletoe were used and two RCTs did not report the specific extract administered. The synthesis of outcomes of this SR was based on three comparisons relating to the control arm of the RCTs. Five RCTs (72%) compared ME to no additional treatment, and three reported on mortality and recurrence and were able to be included in the meta-analyses of mortality and recurrence. In two RCTs (29%), ME was not used as an adjunctive element to therapy but instead was compared against a common cancer treatment (i.e., etoposide or bacillus Calmette-Guérin (BCG)), wherein the intervention group did not receive standard care. Five RCTs (72%) administered ME subcutaneously, and four out of the seven RCTs (57%) used it during the postoperative period. Duration of therapy in the RCTs ranged from intra-operatively only up to over a 60-week period (cycling with breaks). All these variations contribute to the limitation of the results of the current SR.
From the current SR, the effect of ME on mortality and recurrence, including metastasis, was inconsistent, and the evidence of these outcomes needs to be explored in future high-quality research studies. While meta-analysis for mortality and recurrence revealed no significant difference between MEs and no added treatment [68,70,75], one small RCT in osteosarcoma patients, Longhi et al., 2009, reported favoring but not significant relapse and survival results at 12 months using ME versus etoposide [71]. The increased relapse rate and disease progression in an RCT that administered intravesical ME in superficial bladder cancer patients versus BCG was attributed to the relatively insufficient dose of ME used [69]. In contrast, there were some positive effects on quality of life and immune and inflammatory markers from the application of ME in three comparisons (no added treatment, colorectal patients, and active treatments) [15,71,73,74].
The details of adverse events were poorly reported across the RCTs. Overall, there were no significant concerning side effects, serious or severe adverse events, and/or postoperative complications from using MEs in cancer patients perioperatively [68,69,74]. One study even reported a better overall side effect profile with the use of MEs [71]. However, it is important to note that two RCTs highlighted concerns that were related to the application of MEs compared to no added interventions [70,75]. Kleeberg et al., 2004, reported that among patients with melanoma, treatment discontinuation due to grade 3–4 toxicities was reported in 5 out of 102 (5%) patients who received Iscador® M in comparison to 0 out of 102 (0%) patients in the no added treatment group [70]. Similarly, Steuer-Vogt et al., 2001, in addition to the reported local and systemic side effects resulting from mistletoe SC injections into the abdominal wall, reported 16 patients who received Eurixor® (mistletoe extract) who refused further injections because of mistletoe-related AEs (without a precise definition of these AEs) [75]. One controlled cohort study conducted by Augustin in 2005 evaluating 329 participants who had received mistletoe extract for a median of 30 months reported 11 (3%) systemic adverse drug reactions (ADRs) attributed to the mistletoe treatment, 42 local injection site reactions, and 6 mistletoe treatment terminations due to ADRs [77]. No safety concerns overall were identified from the non-randomized and observational trials [76,77,78].
There is a paucity of data from other systematic reviews specifically reporting on the efficacy and safety of using MEs in oncology patients during the perioperative period; moreover, the use of MEs among oncology patients in general is still an area of controversy. There are many reports on MEs used alongside conventional and standard treatment in oncology patients. Most of these RCTs evaluated quality of life, symptom management, and side effects from standard treatment and reported positive effects from the use of MEs for these outcomes [15,72,79,80,81,82,83,84,85,86,87]. These results are also supported by a recent SR [88]. Survival results across studies were mixed; some reported benefits [72,73], and some did not [84,89]. The SRs of the clinical trials reported methodological flaws regarding the included RCTs, making the interpretation of results challenging [2,90,91,92,93,94]. In addition, there is some controversy about the results of some of these SRs [92,93,95,96,97]. In general, the methodological quality appraisals of our SR revealed a substantial risk of bias across all included RCTs with none judged to have a low risk of bias overall (Figure 2, Table 3 and Table 4). The high risk of bias is consistent with reports from other SRs [88,93,94]. Methodological quality varied considerably between the non-randomized studies in the current SR, with the Augustin study [77] scoring best with a quality score of 9 out of 9 according to the NOS. However, the “Outcome” and “comparability of cohorts on the basis of the design or analysis” domains demonstrated several limitations and inadequacy, respectively, in the Bussing and Elsasser-Beile studies [76,78].
The strengths of the current SR and MA include being the first SR to address a particular oncology population (perioperative oncology patients) and examine the use of MEs among them, comprehensiveness of the review process, providing an up-to-date review of the literature, a priori protocol registration, and transparency in reporting the outcomes. Furthermore, all the steps of this SR were duplicated to ensure accuracy. In addition, the inclusion of the non-randomized trials for safety data and information regarding the application of MEs in cancer patients during the perioperative period strengthened the review [98,99]. The main limitation of the methodology was in excluding non-English reports and not searching Asian databases, which introduced a language bias.
The perioperative period in cancer patients is challenging as it usually includes major surgeries, stressful and lengthy preparations, hospital admission, and prolonged recovery [98]. Surgically induced stress, the resultant inflammatory response, and a weakened immune response might promote angiogenesis, tumor shedding and metastasis, and proliferation of tumor growth factors [100]. Aiming for a favorable immune balance during the perioperative period is essential for long-term benefits, including the survival of cancer patients [101]. One strategy to overcome these challenges and achieve the immune balance is combining cancer surgery and immunotherapy [100]. However, as a complex treatment strategy, patients may develop more pronounced side effects and limitations before having any benefit. Furthermore, immunomodulation may have adverse effects on wound healing, recovery, and risk of infection, and the benefits, especially among cancer patients, need to be weighed against the risks [100].
Despite the limitations of the evidence we have reviewed, there is a signal of positive effects from using MEs during the perioperative period in cancer patients, in particular for immune support and quality of life. There is sufficient evidence to call for more research based on the recent guidelines in this area, in particular for colorectal cancer during the perioperative period. The low risk of side effects seen from the available research and the potential for reduced immunosuppression, in addition to MEs’ cytotoxic effects, provide a compelling basis for further evaluation [4,19,21,22,23,24,25,26].

5. Conclusions

Our systematic review suggests a possible benefit of using ME with respect to positive impacts on the immune system (especially NK cells), and also potentially on quality of life, in patients with cancer during the perioperative period. With respect to survival, results showed no evidence of a difference compared to no added therapy and were inconsistent. Nevertheless, the evidence base is fairly sparse due to different outcomes evaluated across studies, and has methodological limitations rated as either unclear or high risk of bias. Further high-quality research is warranted to investigate the use of ME in the perioperative setting, especially in patients with colorectal cancer.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/curroncol30090595/s1, MEDLINE Search Strategy, Table S1: Characteristics of the 3 nonrandomized and observational Studies, Table S2: Interventions/exposures in the 3 nonrandomized and observational studies, Table S3: Summary characteristics of 7 RCTs and 3 non-RCTs, Table S4: Additional results from RCTs, Table S5: Results from nonrandomized and observational studies.

Author Contributions

Conceptualization, P.P.; Methodology, E.C., P.P. and M.E.; Formal Analysis, E.C.; Investigation, E.C., M.E., P.P., K.C. and D.S.; Resources, P.P., K.C. and D.S.; Data Curation, E.C., M.E., V.L., K.C., C.A., S.B. and A.P.; Writing—Original Draft Preparation, E.C., M.E., D.S, K.C., S.B., A.P., P.P., C.A. and V.L.; Visualization, E.C.; Supervision, P.P., K.C. and D.S.; Funding Acquisition, P.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Patterson Institute for Integrative Oncology Research at the Canadian College of Naturopathic Medicine.

Institutional Review Board Statement

Not applicable. No primary research was conducted in this synthesis project.

Informed Consent Statement

Not applicable. No primary research was conducted.

Data Availability Statement

The data presented in this study are available in detail in the online Supplementary Material for this article.

Acknowledgments

The authors wish to sincerely thank John and Thea Patterson, Jessie McGowan, Nancy Rawling.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kienle, G.S.; Grugel, R.; Kiene, H. Safety of higher dosages of Viscum album L. in animals and humans—Systematic review of immune changes and safety parameters. BMC Complement. Altern. Med. 2011, 11, 72. [Google Scholar] [CrossRef] [PubMed]
  2. Kienle, G.S.; Kiene, H. Review article: Influence of Viscum album L. (European mistletoe) extracts on quality of life in cancer patients: A systematic review of controlled clinical studies. Integr. Cancer Ther. 2010, 9, 142–157. [Google Scholar] [CrossRef] [PubMed]
  3. Mistletoe—Professional Monograph; Unpublished Data; The Center of Health Innovation: Ottawa, ON, Canada, 2022; Available online: https://thechi.ca/research/ (accessed on 17 July 2023).
  4. Szurpnicka, A.; Kowalczuk, A.; Szterk, A. Biological activity of mistletoe: In vitro and in vivo studies and mechanisms of action. Arch. Pharmacal Res. 2020, 43, 593–629. [Google Scholar] [CrossRef] [PubMed]
  5. Bussing, A. Mistletoe: The Genus Viscum; Hardwood Academic Publishers: Amsterdam, The Netherlands, 2000. [Google Scholar]
  6. Lev, E.; Ephraim, M.; Ben-Arye, E. European and Oriental mistletoe: From mythology to contemporary integrative cancer care. Eur. J. Integr. Med. 2011, 3, e133–e137. [Google Scholar] [CrossRef]
  7. Steiner, R. Geisteswissenschaft und Medizin; Rudolf Steiner Verlag: Dornach, Switzerland, 1985. [Google Scholar]
  8. Ostermann, T.; Raak, C.; Büssing, A. Survival of cancer patients treated with mistletoe extract (Iscador): A systematic literature review. BMC Cancer 2009, 9, 451. [Google Scholar] [CrossRef] [PubMed]
  9. Ben-Arye, E.; Attias, S.; Tadmor, T.; Schiff, E. Herbs in hemato-oncological care: An evidence-based review of data on efficacy, safety, and drug interactions. Leuk. Lymphoma 2010, 51, 1414–1423. [Google Scholar] [CrossRef] [PubMed]
  10. Horneber, M.A.; Bueschel, G.; Huber, R.; Linde, K.; Rostock, M. Mistletoe therapy in oncology. Cochrane Database Syst. Rev. 2008, 2008, CD003297. [Google Scholar] [CrossRef]
  11. Büssing, A. Induction of apoptosis by the mistletoe lectins: A review on the mechanisms of cytotoxicity mediated by Viscum album L. Apoptosis 1996, 1, 25–32. [Google Scholar] [CrossRef]
  12. Stein, G.M.; Schaller, G.; Pfüller, U.; Schietzel, M.; Büssing, A. Thionins from Viscum album L: Influence of the viscotoxins on the activation of granulocytes. Anticancer Res. 1999, 19, 1037–1042. [Google Scholar]
  13. Gabius, H.J.; Gabius, S.; Joshi, S.S.; Koch, B.; Schroeder, M.; Manzke, W.M.; Westerhausen, M. From ill-defined extracts to the immunomodulatory lectin: Will there be a reason for oncological application of mistletoe? Planta Med. 1994, 60, 2–7. [Google Scholar] [CrossRef]
  14. Samtleben, R.; Hajto, T.; Hostanska, K.; Wagner, H. Mistletoe lectins as immunostimulants (chemistry, pharmacology and clinic). In Immunomodulatory Agents from Plants; Wagner, H., Ed.; Birkhäuser: Basel, Switzerland, 1999; pp. 223–241. [Google Scholar]
  15. Enesel, M.B.; Acalovschi, I.; Grosu, V.; Sbarcea, A.; Rusu, C.; Dobre, A.; Weiss, T.; Zarkovic, N. Perioperative application of the Viscum album extract Isorel in digestive tract cancer patients. Anticancer Res. 2005, 25, 4583–4590. [Google Scholar] [PubMed]
  16. PDQ Integrative, Alternative, and Complementary Therapies Editorial Board. Mistletoe Extracts (PDQ®): Health Professional Version. In PDQ Cancer Information Summaries; National Cancer Institute (US): Bethesda, MD, USA, 2002.
  17. Hajto, T.; Hostanska, K.; Gabius, H.J. Modulatory potency of the beta-galactoside-specific lectin from mistletoe extract (Iscador) on the host defense system in vivo in rabbits and patients. Cancer Res. 1989, 49, 4803–4808. [Google Scholar] [PubMed]
  18. Beuth, J.; Stoffel, B.; Ko, H.L.; Buss, G.; Tunggal, L.; Pulverer, G. Immunoactive effects of various mistletoe lectin-1 dosages in mammary carcinoma patients. Arzneimittelforschung 1995, 45, 505–507. [Google Scholar] [PubMed]
  19. Park, Y.K.; Do, Y.R.; Jang, B.C. Apoptosis of K562 leukemia cells by Abnobaviscum F®, a European mistletoe extract. Oncol. Rep. 2012, 28, 2227–2232. [Google Scholar] [CrossRef] [PubMed]
  20. Fan, J.; Wu, M.; Wang, J.; Ren, D.; Zhao, J.; Yang, G. 1,7-Bis(4-hydroxyphenyl)-1,4-heptadien-3-one induces lung cancer cell apoptosis via the PI3K/Akt and ERK1/2 pathways. J. Cell. Physiol. 2019, 234, 6336–6349. [Google Scholar] [CrossRef] [PubMed]
  21. Janssen, O.; Scheffler, A.; Kabelitz, D. In vitro effects of mistletoe extracts and mistletoe lectins. Cytotoxicity towards tumor cells due to the induction of programmed cell death (apoptosis). Arzneimittelforschung 1993, 43, 1221–1227. [Google Scholar] [PubMed]
  22. Mishra, R.; Sharma, S.; Sharma, R.S.; Singh, S.; Sardesai, M.M.; Sharma, S.; Mishra, V. Viscum articulatum Burm. f. aqueous extract exerts antiproliferative effect and induces cell cycle arrest and apoptosis in leukemia cells. J. Ethnopharmacol. 2018, 219, 91–102. [Google Scholar] [CrossRef]
  23. Elluru, S.R.; Duong Van Huyen, J.P.; Delignat, S.; Prost, F.; Heudes, D.; Kazatchkine, M.D.; Friboulet, A.; Kaveri, S.V. Antiangiogenic properties of Viscum album extracts are associated with endothelial cytotoxicity. Anticancer Res. 2009, 29, 2945–2950. [Google Scholar]
  24. Olsnes, S.; Stirpe, F.; Sandvig, K.; Pihl, A. Isolation and characterization of viscumin, a toxic lectin from Viscum album L. (mistletoe). J. Biol. Chem. 1982, 257, 13263–13270. [Google Scholar] [CrossRef]
  25. Stirpe, F.; Sandvig, K.; Olsnes, S.; Pihl, A. Action of viscumin, a toxic lectin from mistletoe, on cells in culture. J. Biol. Chem. 1982, 257, 13271–13277. [Google Scholar] [CrossRef]
  26. Büssing, A.; Schaller, G.; Pfüller, U. Generation of reactive oxygen intermediates (ROI) by the thionins from Viscum album L. Anticancer Res. 1998, 18, 4291–4296. [Google Scholar] [PubMed]
  27. Elluru, S.R.; Duong van Huyen, J.P.; Delignat, S.; Kazatchkine, M.D.; Friboulet, A.; Kaveri, S.V.; Bayry, J. Induction of maturation and activation of human dendritic cells: A mechanism underlying the beneficial effect of Viscum album as complimentary therapy in cancer. BMC Cancer 2008, 8, 161. [Google Scholar] [CrossRef] [PubMed]
  28. Kim, J.J.; Hwang, Y.H.; Kang, K.Y.; Kim, I.; Kim, J.B.; Park, J.H.; Yoo, Y.C.; Yee, S.T. Enhanced dendritic cell maturation by the B-chain of Korean mistletoe lectin (KML-B), a novel TLR4 agonist. Int. Immunopharmacol. 2014, 21, 309–319. [Google Scholar] [CrossRef] [PubMed]
  29. Steinborn, C.; Klemd, A.M.; Sanchez-Campillo, A.S.; Rieger, S.; Scheffen, M.; Sauer, B.; Garcia-Käufer, M.; Urech, K.; Follo, M.; Ücker, A.; et al. Viscum album neutralizes tumor-induced immunosuppression in a human in vitro cell model. PLoS ONE 2017, 12, e0181553. [Google Scholar] [CrossRef] [PubMed]
  30. Podlech, O.; Harter, P.N.; Mittelbronn, M.; Pöschel, S.; Naumann, U. Fermented mistletoe extract as a multimodal antitumoral agent in gliomas. Evid.-Based Complement. Altern. Med. 2012, 2012, 501796. [Google Scholar] [CrossRef] [PubMed]
  31. Hajto, T. Immunomodulatory effects of iscador: A Viscum album preparation. Oncology 1986, 43 (Suppl. S1), 51–65. [Google Scholar] [CrossRef] [PubMed]
  32. Hajto, T.; Hostanska, K.; Frei, K.; Rordorf, C.; Gabius, H.J. Increased secretion of tumor necrosis factors alpha, interleukin 1, and interleukin 6 by human mononuclear cells exposed to beta-galactoside-specific lectin from clinically applied mistletoe extract. Cancer Res. 1990, 50, 3322–3326. [Google Scholar] [PubMed]
  33. Kohl, B.A.; Deutschman, C.S. The inflammatory response to surgery and trauma. Curr. Opin. Crit. Care 2006, 12, 325–332. [Google Scholar] [CrossRef]
  34. Aller, M.A.; Arias, J.L.; Nava, M.P.; Arias, J. Posttraumatic inflammation is a complex response based on the pathological expression of the nervous, immune, and endocrine functional systems. Exp. Biol. Med. 2004, 229, 170–181. [Google Scholar] [CrossRef]
  35. Arias, J.I.; Aller, M.A.; Arias, J. Surgical inflammation: A pathophysiological rainbow. J. Transl. Med. 2009, 7, 19. [Google Scholar] [CrossRef]
  36. Dabrowska, A.M.; Slotwinski, R. The immune response to surgery and infection. Cent. Eur. J. Immunol. 2014, 39, 532–537. [Google Scholar] [CrossRef] [PubMed]
  37. Bone, R.C.; Balk, R.A.; Cerra, F.B.; Dellinger, R.P.; Fein, A.M.; Knaus, W.A.; Schein, R.M.; Sibbald, W.J. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Chest 1992, 101, 1644–1655. [Google Scholar] [CrossRef]
  38. Bone, R.C.; Grodzin, C.J.; Balk, R.A. Sepsis: A new hypothesis for pathogenesis of the disease process. Chest 1997, 112, 235–243. [Google Scholar] [CrossRef] [PubMed]
  39. Hildebrand, F.; Pape, H.C.; Krettek, C. The importance of cytokines in the posttraumatic inflammatory reaction. Unfallchirurg 2005, 108, 793–803. [Google Scholar] [CrossRef]
  40. Bonamin, L.V.; de Carvalho, A.C.; Waisse, S. Viscum album (L.) in experimental animal tumors: A meta-analysis. Exp. Ther. Med. 2017, 13, 2723–2740. [Google Scholar] [CrossRef] [PubMed]
  41. Tsuchiya, Y.; Sawada, S.; Yoshioka, I.; Ohashi, Y.; Matsuo, M.; Harimaya, Y.; Tsukada, K.; Saiki, I. Increased surgical stress promotes tumor metastasis. Surgery 2003, 133, 547–555. [Google Scholar] [CrossRef] [PubMed]
  42. Glasner, A.; Avraham, R.; Rosenne, E.; Benish, M.; Zmora, O.; Shemer, S.; Meiboom, H.; Ben-Eliyahu, S. Improving survival rates in two models of spontaneous postoperative metastasis in mice by combined administration of a beta-adrenergic antagonist and a cyclooxygenase-2 inhibitor. J. Immunol. 2010, 184, 2449–2457. [Google Scholar] [CrossRef]
  43. McCrath, D.J.; Cerboni, E.; Frumento, R.J.; Hirsh, A.L.; Bennett-Guerrero, E. Thromboelastography maximum amplitude predicts postoperative thrombotic complications including myocardial infarction. Anesth. Analg. 2005, 100, 1576–1583. [Google Scholar] [CrossRef]
  44. Seth, R.; Tai, L.H.; Falls, T.; de Souza, C.T.; Bell, J.C.; Carrier, M.; Atkins, H.; Boushey, R.; Auer, R.A. Surgical stress promotes the development of cancer metastases by a coagulation-dependent mechanism involving natural killer cells in a murine model. Ann. Surg. 2013, 258, 158–168. [Google Scholar] [CrossRef]
  45. Tartter, P.I.; Steinberg, B.; Barron, D.M.; Martinelli, G. The prognostic significance of natural killer cytotoxicity in patients with colorectal cancer. Arch. Surg. 1987, 122, 1264–1268. [Google Scholar] [CrossRef]
  46. Tai, L.H.; de Souza, C.T.; Belanger, S.; Ly, L.; Alkayyal, A.A.; Zhang, J.; Rintoul, J.L.; Ananth, A.A.; Lam, T.; Breitbach, C.J.; et al. Preventing postoperative metastatic disease by inhibiting surgery-induced dysfunction in natural killer cells. Cancer Res. 2013, 73, 97–107. [Google Scholar] [CrossRef] [PubMed]
  47. Shakhar, G.; Ben-Eliyahu, S. Potential prophylactic measures against postoperative immunosuppression: Could they reduce recurrence rates in oncological patients? Ann. Surg. Oncol. 2003, 10, 972–992. [Google Scholar] [CrossRef] [PubMed]
  48. Li, T.S.; Kaneda, Y.; Ueda, K.; Hamano, K.; Zempo, N.; Esato, K. The influence of tumour resection on angiostatin levels and tumour growth—An experimental study in tumour-bearing mice. Eur. J. Cancer 2001, 37, 2283–2288. [Google Scholar] [CrossRef] [PubMed]
  49. Kumara, H.M.; Feingold, D.; Kalady, M.; Dujovny, N.; Senagore, A.; Hyman, N.; Cekic, V.; Whelan, R.L. Colorectal resection is associated with persistent proangiogenic plasma protein changes: Postoperative plasma stimulates in vitro endothelial cell growth, migration, and invasion. Ann. Surg. 2009, 249, 973–977. [Google Scholar] [CrossRef] [PubMed]
  50. van der Bilt, J.D.; Borel Rinkes, I.H. Surgery and angiogenesis. Biochim. Biophys. Acta 2004, 1654, 95–104. [Google Scholar] [CrossRef] [PubMed]
  51. Bailey, C.E.; Parikh, A.A. Assessment of the risk of antiangiogenic agents before and after surgery. Cancer Treat. Rev. 2018, 68, 38–46. [Google Scholar] [CrossRef] [PubMed]
  52. Stöcker, A.; Mehnert-Theuerkauf, A.; Hinz, A.; Ernst, J. Utilization of complementary and alternative medicine (CAM) by women with breast cancer or gynecological cancer. PLoS ONE 2023, 18, e0285718. [Google Scholar] [CrossRef]
  53. Huebner, J.; Muenstedt, K.; Prott, F.J.; Stoll, C.; Micke, O.; Buentzel, J.; Muecke, R.; Senf, B. Online survey of patients with breast cancer on complementary and alternative medicine. Breast Care 2014, 9, 60–63. [Google Scholar] [CrossRef]
  54. Grimm, D.A.-O.; Voiss, P.; Paepke, D.; Dietmaier, J.; Cramer, H.; Kümmel, S.; Beckmann, M.W.; Woelber, L.; Schmalfeldt, B.; Freitag, U.; et al. Gynecologists’ attitudes toward and use of complementary and integrative medicine approaches: Results of a national survey in Germany. Arch. Gynecol. Obstet. 2021, 303, 967–980. [Google Scholar] [CrossRef]
  55. Bussing, A.; Raak, C.; Ostermann, T. Quality of life and related dimensions in cancer patients treated with mistletoe extract (iscador): A meta-analysis. Evid.-Based Complement. Altern. Med. 2012, 2012, 219402. [Google Scholar] [CrossRef]
  56. Beuth, J. Evidence-based complementary oncology: Innovative approaches to optimise standard therapy strategies. Anticancer Res. 2010, 30, 1767–1771. [Google Scholar] [PubMed]
  57. Cogo, E.; Papadogianis, P. Characteristics of 218 Recent Reviews on Natural Health Products in Integrative Cancer Care: A bibliometric analysis of trends in the human research literature. J. Orthomol. Med. 2018, 33, 1–13. [Google Scholar]
  58. Cogo, E.; Elsayed, M.; Liang, V.; Cooley, K.; Guerin, C.; Psihogios, A.; Papadogianis, P. Are Supplemental Branched-Chain Amino Acids Beneficial during the Oncological Peri-Operative Period: A Systematic Review and Meta-Analysis. Integr. Cancer Ther. 2021, 20, 1534735421997551. [Google Scholar] [CrossRef] [PubMed]
  59. Cogo, E.; Elsayed, M.; Liang, V.; Cooley, K.; Guerin, C.; Psihogios, A.; Papadogianis, P. Probiotics Evaluation in Oncological Surgery: A Systematic Review of 36 Randomized Controlled Trials Assessing 21 Diverse Formulations. Curr. Oncol. 2021, 28, 5192–5214. [Google Scholar] [CrossRef] [PubMed]
  60. Higgins, J.P.T.; Thomas, J.; Chandler, J.; Cumpston, M.; Li, T.; Page, M.J.; Welch, V.A.E. Cochrane Handbook for Systematic Reviews of Interventions; Version 6.2 (Updated February 2021); Cochrane: London, UK, 2021; Available online: www.training.cochrane.org/handbook (accessed on 1 September 2021).
  61. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009, 6, e1000097. [Google Scholar] [CrossRef] [PubMed]
  62. Page, M.A.-O.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  63. McGowan, J.; Sampson, M.; Salzwedel, D.M.; Cogo, E.; Foerster, V.; Lefebvre, C. PRESS Peer Review of Electronic Search Strategies: 2015 Guideline Statement. J. Clin. Epidemiol. 2016, 75, 40–46. [Google Scholar] [CrossRef]
  64. Higgins, J.P.; Altman, D.G.; Gøtzsche, P.C.; Jüni, P.; Moher, D.; Oxman, A.D.; Savovic, J.; Schulz, K.F.; Weeks, L.; Sterne, J.A. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ 2011, 343, d5928. [Google Scholar] [CrossRef]
  65. Wells, G.A.; Shea, B.; O’Connell, D.; Peterson, J.; Welch, V.; Losos, M.; Tugwell, P. The Newcastle-Ottawa Scale (NOS) for Assessing the Quality of Nonrandomised Studies in Meta-Analyses. Available online: http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp (accessed on 1 September 2021).
  66. STATA, version 12; Computer Program; StataCorp LLC: College Station, TX, USA, 2011.
  67. CIOMS Working Group X. Evidence Synthesis and Meta-Analysis for Drug Safety: Report of CIOMS Working Group X; Council for International Organizations of Medical Sciences (CIOMS): Geneva, Switzerland, 2016; Available online: https://cioms.ch/publications/product/evidence-synthesis-and-meta-analysis-report-of-cioms-working-group-x/ (accessed on 1 September 2021).
  68. Goebell, P.J.; Otto, T.; Suhr, J.; Rübben, H. Evaluation of an unconventional treatment modality with mistletoe lectin to prevent recurrence of superficial bladder cancer: A randomized phase II trial. J. Urol. 2002, 168, 72–75. [Google Scholar] [CrossRef]
  69. Ibrahiem, E.; Hekal, I. UP-3.040: Mistletoe Extract as an Adjuvant Therapy after Resection of Superficial Bladder Cancer: Prospective Clinical Randomized Study. Urology 2009, 74, S306. [Google Scholar] [CrossRef]
  70. Kleeberg, U.R.; Suciu, S.; Bröcker, E.B.; Ruiter, D.J.; Chartier, C.; Liénard, D.; Marsden, J.; Schadendorf, D.; Eggermont, A.M. Final results of the EORTC 18871/DKG 80-1 randomised phase III trial. rIFN-alpha2b versus rIFN-gamma versus ISCADOR M versus observation after surgery in melanoma patients with either high-risk primary (thickness > 3 mm) or regional lymph node metastasis. Eur. J. Cancer 2004, 40, 390–402. [Google Scholar] [CrossRef] [PubMed]
  71. Longhi, A.; Mariani, E.; Kuehn, J.J. A randomized study with adjuvant mistletoe versus oral Etoposide on post relapse disease-free survival in osteosarcoma patients. Eur. J. Integr. Med. 2009, 1, 27–33. [Google Scholar] [CrossRef]
  72. Longhi, A.; Reif, M.; Mariani, E.; Ferrari, S. A Randomized Study on Postrelapse Disease-Free Survival with Adjuvant Mistletoe versus Oral Etoposide in Osteosarcoma Patients. Evid.-Based Complement. Altern. Med. 2014, 2014, 210198. [Google Scholar] [CrossRef]
  73. Longhi, A.; Cesari, M.; Serra, M.; Mariani, E. Long-Term Follow-up of a Randomized Study of Oral Etoposide versus Viscum album Fermentatum Pini as Maintenance Therapy in Osteosarcoma Patients in Complete Surgical Remission after Second Relapse. Sarcoma 2020, 2020, 8260730. [Google Scholar] [CrossRef] [PubMed]
  74. Schink, M.; Tröger, W.; Dabidian, A.; Goyert, A.; Scheuerecker, H.; Meyer, J.; Fischer, I.U.; Glaser, F. Mistletoe extract reduces the surgical suppression of natural killer cell activity in cancer patients. a randomized phase III trial. Forsch. Komplementärmedizin 2007, 14, 9–17. [Google Scholar] [CrossRef] [PubMed]
  75. Steuer-Vogt, M.K.; Bonkowsky, V.; Ambrosch, P.; Scholz, M.; Neiss, A.; Strutz, J.; Hennig, M.; Lenarz, T.; Arnold, W. The effect of an adjuvant mistletoe treatment programme in resected head and neck cancer patients: A randomised controlled clinical trial. Eur. J. Cancer 2001, 37, 23–31. [Google Scholar] [CrossRef] [PubMed]
  76. Elsässer-Beile, U.; Leiber, C.; Wetterauer, U.; Bühler, P.; Wolf, P.; Lucht, M.; Mengs, U. Adjuvant intravesical treatment with a standardized mistletoe extract to prevent recurrence of superficial urinary bladder cancer. Anticancer Res. 2005, 25, 4733–4736. [Google Scholar]
  77. Augustin, M.; Bock, P.R.; Hanisch, J.; Karasmann, M.; Schneider, B. Safety and efficacy of the long-term adjuvant treatment of primary intermediate- to high-risk malignant melanoma (UICC/AJCC stage II and III) with a standardized fermented European mistletoe (Viscum album L.) extract. Results from a multicenter, comparative, epidemiological cohort study in Germany and Switzerland. Arzneimittelforschung 2005, 55, 38–49. [Google Scholar] [CrossRef]
  78. Büssing, A.; Bischof, M.; Hatzmann, W.; Bartsch, F.; Soto-Vera, D.; Fronk, E.M.; Gmeindl, M.; Stein, G.M. Prevention of surgery-induced suppression of granulocyte function by intravenous application of a fermented extract from Viscum album L. in breast cancer patients. Anticancer Res. 2005, 25, 4753–4757. [Google Scholar]
  79. Tröger, W.; Zdrale, Z.; Tišma, N.; Matijašević, M. Additional Therapy with a Mistletoe Product during Adjuvant Chemotherapy of Breast Cancer Patients Improves Quality of Life: An Open Randomized Clinical Pilot Trial. Evid.-Based Complement. Altern. Med. 2014, 2014, 430518. [Google Scholar] [CrossRef]
  80. Pelzer, F.; Tröger, W.; Nat, D.R. Complementary Treatment with Mistletoe Extracts During Chemotherapy: Safety, Neutropenia, Fever, and Quality of Life Assessed in a Randomized Study. J. Altern. Complement. Med. 2018, 24, 954–961. [Google Scholar] [CrossRef] [PubMed]
  81. Kim, K.C.; Yook, J.H.; Eisenbraun, J.; Kim, B.S.; Huber, R. Quality of life, immunomodulation and safety of adjuvant mistletoe treatment in patients with gastric carcinoma—A randomized, controlled pilot study. BMC Complement. Altern. Med. 2012, 12, 172. [Google Scholar] [CrossRef] [PubMed]
  82. Tröger, W.; Jezdić, S.; Zdrale, Z.; Tišma, N.; Hamre, H.J.; Matijašević, M. Quality of life and neutropenia in patients with early stage breast cancer: A randomized pilot study comparing additional treatment with mistletoe extract to chemotherapy alone. Breast Cancer 2009, 3, 35–45. [Google Scholar] [CrossRef] [PubMed]
  83. Semiglazov, V.F.; Stepula, V.V.; Dudov, A.; Schnitker, J.; Mengs, U. Quality of life is improved in breast cancer patients by Standardised Mistletoe Extract PS76A2 during chemotherapy and follow-up: A randomised, placebo-controlled, double-blind, multicentre clinical trial. Anticancer Res. 2006, 26, 1519–1529. [Google Scholar] [PubMed]
  84. Bar-Sela, G.; Haim, N. Abnoba-viscum (mistletoe extract) in metastatic colorectal carcinoma resistant to 5-fluorouracil and leucovorin-based chemotherapy. Med. Oncol. 2004, 21, 251–254. [Google Scholar] [CrossRef] [PubMed]
  85. Piao, B.K.; Wang, Y.X.; Xie, G.R.; Mansmann, U.; Matthes, H.; Beuth, J.; Lin, H.S. Impact of complementary mistletoe extract treatment on quality of life in breast, ovarian and non-small cell lung cancer patients. A prospective randomized controlled clinical trial. Anticancer Res. 2004, 24, 303–309. [Google Scholar] [PubMed]
  86. Tröger, W.; Galun, D.; Reif, M.; Schumann, A.; Stanković, N.; Milićević, M. Quality of life of patients with advanced pancreatic cancer during treatment with mistletoe: A randomized controlled trial. Dtsch. Ärztebl. Int. 2014, 111, 493–502. [Google Scholar] [CrossRef]
  87. Beuth, J.; Schneider, B.; Schierholz, J.M. Impact of complementary treatment of breast cancer patients with standardized mistletoe extract during aftercare: A controlled multicenter comparative epidemiological cohort study. Anticancer Res. 2008, 28, 523–527. [Google Scholar]
  88. Loef, M.; Walach, H. Quality of life in cancer patients treated with mistletoe: A systematic review and meta-analysis. BMC Complement. Med. Ther. 2020, 20, 227. [Google Scholar] [CrossRef]
  89. Mansky, P.J.; Wallerstedt, D.B.; Sannes, T.S.; Stagl, J.; Johnson, L.L.; Blackman, M.R.; Grem, J.L.; Swain, S.M.; Monahan, B.P. NCCAM/NCI Phase 1 Study of Mistletoe Extract and Gemcitabine in Patients with Advanced Solid Tumors. Evid.-Based Complement. Altern. Med. 2013, 2013, 964592. [Google Scholar] [CrossRef]
  90. Kienle, G.S.; Berrino, F.; Büssing, A.; Portalupi, E.; Rosenzweig, S.; Kiene, H. Mistletoe in cancer—A systematic review on controlled clinical trials. Eur. J. Med. Res. 2003, 8, 109–119. [Google Scholar] [PubMed]
  91. Kienle, G.S.; Kiene, H. Complementary cancer therapy: A systematic review of prospective clinical trials on anthroposophic mistletoe extracts. Eur. J. Med. Res. 2007, 12, 103–119. [Google Scholar] [PubMed]
  92. Freuding, M.; Keinki, C.; Kutschan, S.; Micke, O.; Buentzel, J.; Huebner, J. Mistletoe in oncological treatment: A systematic review: Part 2: Quality of life and toxicity of cancer treatment. J. Cancer Res. Clin. Oncol. 2019, 145, 927–939. [Google Scholar] [CrossRef] [PubMed]
  93. Freuding, M.; Keinki, C.; Micke, O.; Buentzel, J.; Huebner, J. Mistletoe in oncological treatment: A systematic review: Part 1: Survival and safety. J. Cancer Res. Clin. Oncol. 2019, 145, 695–707. [Google Scholar] [CrossRef] [PubMed]
  94. Ostermann, T.; Appelbaum, S.; Poier, D.; Boehm, K.; Raak, C.; Büssing, A. A Systematic Review and Meta-Analysis on the Survival of Cancer Patients Treated with a Fermented Viscum album L. Extract (Iscador): An Update of Findings. Complement. Med. Res. 2020, 27, 260–271. [Google Scholar] [CrossRef] [PubMed]
  95. Matthes, H.; Hofheinz, R.D.; Bar-Sela, G.; Galun, D.; Martin, D.; Huber, R.; Langhorts, J.; Matthiessen, P.F.; Schad, F. Letter to the editors of the Journal of Cancer Research and Clinical Oncology. J. Cancer Res. Clin. Oncol. 2019, 145, 2405–2407. [Google Scholar] [CrossRef] [PubMed]
  96. Matthes, H.; Thronicke, A.; Hofheinz, R.D.; Baars, E.; Martin, D.; Huber, R.; Breitkreuz, T.; Bar-Sela, G.; Galun, D.; Schad, F. Statement to an Insufficient Systematic Review on Viscum album L. Therapy. Evid.-Based Complement. Altern. Med. 2020, 2020, 7091039. [Google Scholar] [CrossRef]
  97. Huebner, J.; Freuding, M.; Keinki, C.; Micke, O.; Buentzel, J. Answer to the letter to the editors by Matthes and colleagues regarding our systematic reviews on mistletoe. J. Cancer Res. Clin. Oncol. 2019, 145, 2409–2410. [Google Scholar] [CrossRef]
  98. Ackerman, R.S.; Muncey, A.R.; Aldawoodi, N.N.; Kotha, R.; Getting, R.E.G. Cancer Immunotherapies: What the Perioperative Physician Needs to Know. Curr. Oncol. Rep. 2022, 24, 399–414. [Google Scholar] [CrossRef]
  99. Qiu, B.; Cai, K.; Chen, C.; Chen, J.; Chen, K.N.; Chen, Q.X.; Cheng, C.; Dai, T.Y.; Fan, J.; Fan, Z.; et al. Expert consensus on perioperative immunotherapy for local advanced non-small cell lung cancer. Transl. Lung Cancer Res. 2021, 10, 3713–3736. [Google Scholar] [CrossRef]
  100. Bakos, O.; Lawson, C.; Rouleau, S.; Tai, L.H. Combining surgery and immunotherapy: Turning an immunosuppressive effect into a therapeutic opportunity. J. Immunother. Cancer 2018, 6, 86. [Google Scholar] [CrossRef] [PubMed]
  101. Matzner, P.; Sandbank, E.; Neeman, E.; Zmora, O.; Gottumukkala, V.; Ben-Eliyahu, S. Harnessing cancer immunotherapy during the unexploited immediate perioperative period. Nat. Rev. Clin. Oncol. 2020, 17, 313–326. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Study flow diagram.
Figure 1. Study flow diagram.
Curroncol 30 00595 g001
Figure 2. Aggregate risk of bias across studies. Green = low risk; yellow = unclear risk; red = high risk of bias.
Figure 2. Aggregate risk of bias across studies. Green = low risk; yellow = unclear risk; red = high risk of bias.
Curroncol 30 00595 g002
Figure 3. Forest plot of RCTs meta-analysis on mortality, Goebell 2002 [68], Kleeberg 2004 [70], Steuer-Vogt 2001 [75].
Figure 3. Forest plot of RCTs meta-analysis on mortality, Goebell 2002 [68], Kleeberg 2004 [70], Steuer-Vogt 2001 [75].
Curroncol 30 00595 g003
Figure 4. Forest plot of RCTs meta-analysis on recurrence, Goebell 2002 [68], Kleeberg 2004 [70], Steuer-Vogt 2001 [75].
Figure 4. Forest plot of RCTs meta-analysis on recurrence, Goebell 2002 [68], Kleeberg 2004 [70], Steuer-Vogt 2001 [75].
Curroncol 30 00595 g004
Table 1. Characteristics of included RCTs.
Table 1. Characteristics of included RCTs.
Study
Author,
Year
(Study
Period)
CountrySample Size aFundingSurgical Period of Mistletoe
Exposure b
Post-op Mistletoe Initiation Timing bCancer TypesCancer SeverityAge
(y, var.)
%
Female
Adjunct tx
(%)
Schink, 2007
(2002–2004) [74]
Germany32publicIntra-operativeNAColorectal cancerprimary and locally relapsed, stages II–IV71
(NR)
45Antibiotic
(NR)
Enesel,
2005 (NR) [15]
Romania70NRMixed periodsNADigestive tract cancers (51% colorectal)NRNR
(NR)
43NR
Kleeberg, 2004
(1988–2003) [70] c
13 countries: Germany; France; Switzerland; Austria; Belgium; Great Britain; Yugoslavia; Israel; Czechia; Estonia; Spain; Greece; Poland204mixed public and privatePost-operativewithin 6 weeksmelanomaHigh-risk stage IIb (thickness > 3 mm) and stage III (positive lymph nodes) without distant metastasis (49% were Stage Iib)52
(range 14–84)
41NR
Goebell, 2002
(NR) [68]
Germany45publicPost-operative2 weeks postopbladder cancerpTa G1–265 d
(NR)
27NR
Ibrahiem, 2009
(2006–2008) [69] e
Egypt60nonePost-operative1–2 weeks postopSuperficial bladder cancerTa or T1; 82% were Grade 256.1 d
(NR)
NRNR
Steuer-Vogt, 2001
(1993–2000) [75] f
Germany202publicMixed periods fNAHead and neck cancersAll stages58
(range 30–70)
9Antibiotic
(88%)
Longhi, 2020 g
(2007–2019) [73] g
Italy20privatePost-operativemean 1.9 (range 0.7–5.9) monthsOsteosarcoma2nd relapse in the lung (stages I–III)33.9
(range 11–65)
45NR
a Number of patients randomized. b Indicate when mistletoe was started after surgery. c The study design included 2 trials together with 4 total arms but reported mistletoe results versus the control group only (not including the recombinant interferon-alpha and -gamma (rIFN-α2b and IFN-γ) arms. Study age is a median and pertains to all study arms combined. Not all patients followed up for 11 years (due to trial ending and lost to follow-up), but the median follow-up of 8.2 years was for all arms combined in the 2 trials (NR for just the mistletoe trial). d Age reported by median. e Conference abstract. f Mistletoe was started before surgery (1 ± 4 days before surgery), which was achieved in 74% of patients. Stratum A: Surgery-only patients (Data was only extracted from Stratum A). Stratum B: Higher-stage head and neck cancer patients who received surgery and postoperative radiotherapy. g Study closed early in 2011 due to insufficient recruitment. Only the RCT portion of the trial over the 1st 12 months was extracted from all reports (the longer follow-up is more like an observational study due to multiple protocol changes). This also includes the other reports Longhi 2014 and Longhi 2009. Abbreviations: NA = not applicable, NR = not reported, tx = treatment, var = variance, y = year.
Table 2. Interventions and comparators evaluated in the RCTs.
Table 2. Interventions and comparators evaluated in the RCTs.
Study Author, YearGroup/
Brand Name
Characteristics of the Intervention and ControlDosage FrequencyRoute of AdminTx
Duration Pre-op
Tx Duration Post-opTx Duration TOTAL
Schink 2007 [74]Iscador® M special (Weleda, Schwäbisch Gmünd, Germany)Mistletoe extract, 50 mg, IOonceIVNANAOver 1 h
Controlno added tx
Enesel 2005 [15]Isorel® A a
(V. album grown on abietis extract)
Mistletoe extract, 2 vials (60 mg/mL, but the volume per vial was not reported).
Protocol: in the 1st pre-operative week, 1 vial, then 2 vials, then 3 vials every second day and in the 2nd pre-operative week 3, then 2, then 1 vial; and postoperative treatments were repeated in the same manner.
3 × per weekSC224 weeks
Controlno added tx
Kleeberg 2004 [70]Iscador® M (Viscum, mali extract)Mistletoe extract, the dose and the unit were not mentioned.
Protocol was started at dose 0 and then was escalated every other day over 2 weeks starting from 0.01 to reach to 1.0 mg/mL, followed by a 3 day break. Treatment then resumed with 14 doses of 20 mg/mL over 28 days, followed by 7 days of no treatment. Volume per dose NR, but presumption is that 1 mL was given per dose.
3 × per weekInjectionNA1212 months or until tumour progression
Controlno added tx
Goebell 2002 [68]V. album lectin extract (brand name NR)Standardized to mistletoe lectin I (galactoside-specific lectin), 1 mL2 × weekSCNA9 months, 2 cycles of 3 months of treatment separated by a 3-month break9 months
Controlno added tx
Steuer-Vogt 2001 [75]Eurixor®
(biosyn Arzneimittel GmbH,
Fellbach,
Germany)
(mistletoe extract)
Mistletoe lectin-1 (ML-1), 1 ng of ML-1 per kg body weight2 × weekSC1–4 days60 weeks60 weeks b
Controlno added tx
Ibrahiem 2009 [69]Mistletoe extract (brand name was NR)Lectin concentration, 10.000 ng cNRIntravesicalNANRNR
ControlBacillus Calmette–Guérin (BCG), standard therapy cNRIntravesical dNANRNR
Longhi 2020 [73], Longhi 2014 [72], Longhi 2009 [71] fIscador® P (V. album fermentatum Pini extract)Mistletoe, 20 mg e3 × per weekSCNA12 months12 months
ControlEtoposide,
50 mg/m2
QDoralNA6 months; 3 weeks per cycle, 6 cycles, separated by 1-week break6 months
a abietis—firtree; Viscum album extract by Novipharm GmbH, Pörtschach, Austria” was applied in this trial. The fresh mistletoe plant grown on firtree was used to prepare the extract by cold water extraction (60 mg of the whole plant for 1 mL of water). No homogenization or fermentation was applied. Extracted dose is an average. b Mistletoe extract was given in cycles, each consisting of 12 weeks, followed by a break of 4 weeks between 1 and 2, 2 and 3, and 3 and 4 cycles. c Course and dose were given according to the prescribed regimen (prescribed regimen was not mentioned in the abstract). d Standard treatment, given according to prescribed regimen, details of the injection were not reported for the BCG. e The protocol consisted of a starting dose; 2 boxes of series 0 (14 vials, seven vials in each box; 2 vials for each of 0.01 mg and 0.1 mg doses, and three vials of 1 mg), then two boxes of series I; 14 vials (2 vials of each of 0.1 mg, and 1 mg doses, and three vials of 10 mg dose per each box). Further treatment with series II (1, 10, and 20 mg) was given until the 12th month. The site of injection was the abdomen. f All these three citations were on the same patient cohort undergoing the study but with different outcomes. Abbreviations: NA = not applicable, NR = not reported, post-op = postoperatively, Pre-op = pre-operatively, IO = intra-operative, IV = intravenous, SC = subcutaneous, tx = treatment.
Table 3. Cochrane risk of bias appraisal of each RCT a.
Table 3. Cochrane risk of bias appraisal of each RCT a.
Author YearRandom
Sequence Generation (Selection Bias)
Allocation Concealment (Selection Bias)Blinding of Participants and
Personnel (Performance Bias)
Blinding of Outcome
Assessment (Detection Bias)
Incomplete Outcome Data
(Attrition Bias)
Selective
Reporting (Reporting Bias)
Other Bias
Enesel 2005 [15]??--??+
Goebell 2012 [68]+++++?+
Ibrahiem 2009 [69]????+??
Kleeberg 2004 [70]++???++
Longhi 2020 [73]++--++?
Schink 2007 [74]++-?+?+
Steuer-Vogt 2001 [75]++-?+?+
a Green “ + ” = low risk; yellow “?” = unclear risk; red “-” = high risk of bias.
Table 4. NOS quality assessment of non-RCTs a.
Table 4. NOS quality assessment of non-RCTs a.
Study Author, YearSelectionComparabilityOutcome
Augustin, 2005 [77]★★★★★★★★★
Bussing, 2005 [78]★★★★★
Elsasser-Beile, 2005 [76]★★★
a Based on the Newcastle–Ottawa Scale scoring guide, the stars indicate that one of the required quality elements was judged as being present, so a higher star rating (out of 9 maximum) represents higher methodological quality.
Table 5. Other complications (OC) and adverse events in RCTs.
Table 5. Other complications (OC) and adverse events in RCTs.
Study
Author,
Year
Tx
Name
Sample
Size
Follow
-Up
Time
and Unit
All Adverse Effects
(AEs)/Most Common AEs
(N)
(Definition)
Serious AEs
(N)
(Definition
Severe AEs
(N)
(Definition)
Surgical
Complications
(N)
(Definition)
Drug
Reduction
/Treatment
Discontinuation
Number
(Definition)
OC
(N)
(Definition)
Longhi,
2014 [72]
Mistletoe912
mon.
(16)
(definition NR)
(2)
(post
-operative
hospitalization)
(5)
(definition NR)
NR
(NA)
(0)
Dose reduced
(definition NR)

(2)
Medication
discontinued
(definition NR)

(1)
Medication
continued after
interruption
(definition NR)
(2)
Adverse
drug reactions
(Local
erythema
and
hypotension
(1))
Etoposide1012
mon.
(69)
(definition NR)
(3)
(hospitalizations
due to surgery (1)
and
pneumonia a (2)).

3
(26)
(definition NR)
NR
(NA)
(5)
Dose reduced
(definition NR)

(4)
Medication
discontinued
(definition NR)

(18)
Medication
continued after
interruption
(definition NR)
(47)
Adverse
drug
reactions
(Including
G2, G3
hematologic
toxicity,
G-CSF was
necessary
in 3 patients,
1 needed
blood
transfusion
for
G4 anemia.
Most
frequent
ADRs
were
neutropenia,
anemia,
leukopenia,
nausea,
alopecia)
Schink,
2007 [74]
Iscador®
M
special
(mistletoe
extract)
117
days
NR(0)
(definition NR )
NR(4)
(AEs
during
surgery
(hypotension
(2),
hypertension,
circulatory
instability)

(5)
(post-op AEs
UTI (2),
erythocytouria,
body temp
>38 °C
and
C-reactive
protein
>10 mg/dL,
Tension
bulla
beneath
bandage
of
peridural
catheter)
(0)
Allergic
predisposition
to
mistletoe
extract
(hypersensitivity
reaction
to test
injection dose
pre-op
of mistletoe
(0.1 mg)
No added
tx
117
days
NR(1)
(anastomosis
leakage
requiring
2nd surgery)
NR(4)
(AEs
during surgery
hypotension)

(4)
(post-op. AEs
(hypertension
(2),
erythocytouria,
suppuration
of surgical
wound)
NRNR
Enesel,
2005 [15]
No added
tx
30NRNRNRNRNRNRNR
Isorel®
A
(V. album
grown on
abietis
extract)
40NRNRNRNRNRNRNR
Kleeberg,
2004 [70]
Iscador®
M
(Viscum,
mali
extract)
1028
years
NRNRNRNRNR/

(5)
(Tx
discontinuation
d/t
Grade 3–4 toxicity
(WHO
classification))
(0)
organ
toxicity
(definition NR)
No added
tx
1028
years
NRNRNRNRNR/

(0)
(Tx
Discontinuation
d/t
Grade 3–4
toxicity
(WHO
classification))
(0)
organ
toxicity
(definition NR)
Goebell,
2002 [68]
V. album
Lectin
extract
(brand NR)
2318
mon.
(0)
(Adverse
events after
application
of
mistletoe
lectin)
(no specific
Definition)
NRNRNRNRNR
No added
tx
2218
mon.
NRNRNRNRNRNR
Ibrahiem,
2009 [69]
Mistletoe
extract
(brand NR)
30NR(30)
Self-limited
local skin reaction
after
first SC dose b
(definition NR)


(0)
Biochemical
or
hematological
changes
(definition NR)
NRNRNRNRNR
BCG
standard
therapy
30NR(23)
Painful
bladder
sensation
(definition NR)
NRNRNRNRNR
Steuer-
Vogt,
2001 [75]
Eurixor®
(mistletoe
extract)
9760
weeks
(47)
{At start of tx:
pxs with
AEs
upon injections
(local
and/or
systemic
side-effects
upon
mistletoe injections
into
abdominal
wall.
Most
common local
AEs
were rubor
and prurigo)}

(4)
{At 32
weeks or
later:
pxs with
AEs
upon injections
(local
and/or
Systemic
side-effects
upon
mistletoe
injections
into
abdominal
wall}
NRNRNR(16)
Pxs
who refused
further
injections
because
of mistletoe
-induced
AEs
(definition NR)
(4)
Generalized
drug
reactions
(melalgia,
fever ≤ 39 °C,
sleeplessness,
tiredness,
coldness
or heat sensation
and
sneezing)
No added
tx
10560
weeks
NRNRNRNRNRNR
a Pneumonia was regarded as related to etoposide (serious adverse drug reaction). b Intravesical mistletoe was safe and tolerable. Abbreviations: AE = adverse events, mon. = month, N = number, NR = not reported, OC = other complications not already reported on above, pxs = patients, tx = treatment.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cogo, E.; Elsayed, M.; Bhardwaj, S.; Cooley, K.; Aycho, C.; Liang, V.; Papadogianis, P.; Psihogios, A.; Seely, D. Mistletoe Extracts during the Oncological Perioperative Period: A Systematic Review and Meta-Analysis of Human Randomized Controlled Trials. Curr. Oncol. 2023, 30, 8196-8219. https://doi.org/10.3390/curroncol30090595

AMA Style

Cogo E, Elsayed M, Bhardwaj S, Cooley K, Aycho C, Liang V, Papadogianis P, Psihogios A, Seely D. Mistletoe Extracts during the Oncological Perioperative Period: A Systematic Review and Meta-Analysis of Human Randomized Controlled Trials. Current Oncology. 2023; 30(9):8196-8219. https://doi.org/10.3390/curroncol30090595

Chicago/Turabian Style

Cogo, Elise, Mohamed Elsayed, Sukriti Bhardwaj, Kieran Cooley, Christilynn Aycho, Vivian Liang, Peter Papadogianis, Athanasios Psihogios, and Dugald Seely. 2023. "Mistletoe Extracts during the Oncological Perioperative Period: A Systematic Review and Meta-Analysis of Human Randomized Controlled Trials" Current Oncology 30, no. 9: 8196-8219. https://doi.org/10.3390/curroncol30090595

Article Metrics

Back to TopTop