Next Article in Journal
Adult Neurogenesis of the Medial Geniculate Body: In Vitro and Molecular Genetic Analyses Reflect the Neural Stem Cell Capacity of the Rat Auditory Thalamus over Time
Next Article in Special Issue
Advances in HIV Gene Therapy
Previous Article in Journal
HIV Reservoirs and Treatment Strategies toward Curing HIV Infection
Previous Article in Special Issue
CRISPR-Cas9-Based Gene Knockout of Immune Checkpoints in Expanded NK Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

VEGF and Other Gene Therapies Improve Flap Survival—A Systematic Review and Meta-Analysis of Preclinical Studies

by
Wiktor Paskal
1,*,
Mateusz Gotowiec
1,
Albert Stachura
1,2,
Michał Kopka
1,2 and
Paweł Włodarski
1
1
Department of Methodology, Medical University of Warsaw, 1b Banacha Street, 02-091 Warsaw, Poland
2
Doctoral School, Medical University of Warsaw, 81 Żwirki i Wigury Street, 02-091 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(5), 2622; https://doi.org/10.3390/ijms25052622
Submission received: 13 January 2024 / Revised: 12 February 2024 / Accepted: 20 February 2024 / Published: 23 February 2024
(This article belongs to the Special Issue Advances in Gene and Cell Therapy 2.0)

Abstract

:
Surgical flaps are basic tools in reconstructive surgery. Their use may be limited by ischemia and necrosis. Few therapies address or prevent them. Genetic therapy could improve flap outcomes, but primary studies in this field present conflicting results. This systematic review and meta-analysis aimed to appraise the efficacy of external gene delivery to the flap for its survival in preclinical models. This review was registered with PROSPERO (CRD42022359982). PubMed, Embase, Web of Science, and Scopus were searched to identify studies using animal models reporting flap survival outcomes following any genetic modifications. Random-effects meta-analysis was used to calculate mean differences in flap survival with accompanying 95% CI. The risk of bias was assessed using the SYRCLE tool. Subgroup and sensitivity analyses were performed to ascertain the robustness of primary analyses, and the evidence was assessed using the GRADE approach. The initial search yielded 690 articles; 51 were eventually included, 36 of which with 1576 rats were meta-analyzed. VEGF gene delivery to different flap types significantly improved flap survival area by 15.66% (95% CI 11.80–19.52). Other interventions had smaller or less precise effects: PDGF—13.44% (95% CI 3.53–23.35); VEGF + FGF—8.64% (95% CI 6.94–10.34); HGF—5.61% (95% CI 0.43–10.78); FGF 3.84% (95% CI 1.13–6.55). Despite considerable heterogeneity, moderate risk of bias, and low quality of evidence, the efficacy of VEGF gene therapy remained significant in all sensitivity analyses. Preclinical data indicate that gene therapy is effective for increasing flap survival, but further animal studies are required for successful clinical translation.

1. Introduction

Surgical flaps comprise most of the alternatives to the reconstructive ladder. They offer complex tissue defect coverage along with potential muscular, vascular, and neural function donors to the recipient area. Thus, they are often superior to other reconstructive options. Flap harvesting requires sacrificing a tissue bulk from the donor area, and flap survival relies on extensive vascular support in the recipient region. As a result, flaps are prone to postoperative complications (ischemia, venous congestion, and lymphatic drainage dysfunction). Necrosis usually begins distally to the flap pedicle. This is related to insufficient angiogenesis, which depends on cytokines and nutrients delivered from the pedicle. As a result, the distal parts are not only vulnerable to nutrient deprivation and ischemia but also to ischemia-reperfusion injury, which is characterized by the influx of free radicals to nutrient-poor regions shortly after establishing blood flow. Depending on the flap type used, in the clinical settings, 5–20% ischemia or necrosis rate is reported, while preclinical models produce necrosis in 20–80% of the flap area.
Numerous attempts have been made to decrease flap necrosis, particularly in patients with vascular comorbidities who are at a higher risk of revisions and flap loss [1]. Since most flap-based reconstructions are planned surgeries, various preconditioning interventions have been implemented to improve survival. However, effective techniques are limited to surgical delay, local hyperthermia or hypothermia treatment, and pharmacological preconditioning [2]. Intraoperative techniques include the use of vessel supercharging techniques or intraoperative preconditioning. Postoperative ischemia treatment may be addressed by hyperbaric oxygen treatment, local vasodilator application, or topical growth factor treatment [3,4]. Although such interventions can provide short-term increases in vessel density in flap surroundings, they require repetitive administration (except surgical delay) and strongly depend on accessibility to the affected flap tissue. Finally, these techniques do not provide localized support to the distal parts of the flap but rather affect the whole tissue bulk, which can further increase the risk of ischemia-reperfusion injury. Most of them focus on indirect stimulation of angiogenesis. Apart from the surgical delay and hypo/hyperthermia therapy of the panned flap, no other therapies could be implemented before or during flap elevation. Finally, most of the current therapies aim to decrease the area of yet-present necrosis or ischemia.
External delivery of proangiogenic particles is complicated, and repetitive administration methods may increase complication rates. To overcome these limitations, genetic engineering tools have emerged. They allow more localized and specific biological drug delivery, together with long-term target protein production within the tissue [5]. The first report of improved hind limb survival via VEGF-plasmid transfection in a rabbit model was presented by Takeshita in 1996 [6]. Thereafter, a wide variety of target proteins, transformation techniques, and applications in surgical flap animal models have been explored.
To date, there has been no systematic summary of the efficacy of surgical flaps’ genetic modification before, during, or after their elevation. To address this knowledge gap, we performed a systematic review and meta-analysis to assess flap survival outcomes following these interventions in animal models.

2. Results

The initial literature search yielded 690 papers, reduced to 644 after deduplication. After abstract screening, 76 articles were eligible for full-text retrieval. Eventually, 51 studies [7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57] were included in the qualitative synthesis (Table 1), of which 36 studies with 1576 rats were analyzed quantitatively (Figure 1).

2.1. Primary Outcome—Flap Survival

2.1.1. Vascular Endothelial Growth Factor (VEGF)

The authors used VEGF as an intervention in 27 studies and 45 comparisons (Figure 2) [7,8,9,10,11,12,13,15,16,28,29,30,32,33,34,37,38,39,40,41,42,43,45,46,47,48,50]. Eight studies used viruses as vectors, five used cells, and 13 used plasmids. The overall effect estimate for this intervention was a mean increase in flap survival area of 15.66% (95% CI 11.80–19.52), and the results were highly heterogeneous (I2 = 96%).
There were no subgroup differences between the vectors used (χ2 = 2.55, df = 2, p = 0.28), with all subgroups presenting similar heterogeneity. A subgroup analysis based on flap type showed that two thirds of the comparisons studied were performed using McFarlane or epigastric flaps. We detected significant subgroup differences with the highest effect estimate in comparisons using TRAM flap (MD 19.68, 95% CI 10.70–28.65, k = 4 studies, I2 = 56%) and the lowest in musculocutaneous flap (MD 7.41, 95% CI −0.15–14.96, k = 3 comparisons (1 study, I2 = 95%). The overall effect estimate did not change significantly after sensitivity analyses (Figures S1–S5). We performed a subgroup analysis based on the delivery route (Figure S6) and found significant subgroup differences (p < 0.001). To identify potential effect moderators, we used meta-regression considering administration time, assessment time, vector type, and administration route (Table S2); however, the test of moderators was insignificant (p = 0.39), and our model accounted for only 4.27% of total heterogeneity. We did not identify significant publication bias in this group of studies (Figure S7).

2.1.2. Fibroblast Growth Factor (FGF)

This intervention was used in 6 studies and 12 comparisons (Figure 3) [13,17,23,31,39,49]. One study used viruses as vectors, three used plasmids, and two used cells. Interventions with FGF, on average, increased the flap survival by 3.84% (95% CI 1.13–6.55), and there was considerable heterogeneity between studies (I2 = 87%). This effect diminished and became insignificant after sensitivity analyses.
Only the subgroup with a cellular delivery system showed a significant improvement in flap survival (MD 5.99, 95% CI 2.51–9.47). In the two remaining subgroups, improvement was small and insignificant. All studies utilizing a cellular vector had a high risk of bias. Overall, most studies used McFarlane flaps. Flaps’ survival in this group was improved by FGF administration by a mean of 4.65% (95% CI 1.50–7.79)–which was markedly diminished and rendered insignificant after performing sensitivity analyses (Figures S8 and S9). For TRAM and dorsal island skin flaps, FGF did not improve the outcome compared to placebo. We did not identify a significant publication bias in this group of studies (Figure S10).

2.1.3. Platelet-Derived Growth Factor (PDGF)

Platelet-derived growth factor was used in 4 studies and 5 comparisons (Figure 4) [11,17,18,44]. Three studies used plasmids as vectors and one used cells. PDGF treatment increased the flap survival by an average of 13.44% (95% CI 3.53–23.35) with an overall considerable heterogeneity (I2 = 92%). This effect became marginally insignificant (MD 10.94, 95% CI −0.35–22.24) after performing sensitivity analyses (Figures S11 and S12). We only found a significant effect in a subgroup of studies using plasmids (MD 13.75, 95% CI 4.31–23.19) and those utilizing McFarlane flaps (MD 8.78, 95% CI 4.72–12.84). Both effects remained stable and significant after performing the sensitivity analyses (Figures S13–S16).

2.1.4. Hepatocyte Growth Factor (HGF) and HGF + Prostacyclin Synthase (PGIS)

Hepatocyte growth factor alone or enriched with prostacyclin synthase was used in three studies and four comparisons, and one study and four comparisons, respectively (Figure 5) [19,20,53]. Sole HGF treatment increased flap survival by an average of 5.61% (95% CI 0.43–10.78) with moderate heterogeneity (I2 = 52%). An addition of PGIS insignificantly increased flap survival to an average of 8.16% (95% CI −1.09–17.41). Overall, any therapy containing HGF increased flap survival by an average of 6.53% (95% CI 1.59–11.48) with a substantial heterogeneity of included studies (I2 = 64%).

2.1.5. VEGF + FGF

Combined therapy with VEGF and FGF was used in 2 studies and 5 comparisons (Figure 6) [13,49]. On average, it increased flap survival by 8.64% (95% CI 6.94–10.34) with considerable heterogeneity (I2 = 80%).

2.1.6. Other Interventions

Apart from the studies included in the quantitative analysis, we found several studies with various treatment types used in less than three comparisons. Other substances or combinations containing unidentified molecules were used in 15 studies [14,15,21,24,25,26,27,35,36,51,52,54,55,56,57]. The substances or targets previously described include hypoxia-inducible factor, Dikkopf-2, nitric oxide synthase, interleukin-10, angiopoietin, relaxin, NF-κB, KGF, and SDF-1. Studies used a variety of different vectors: plasmids (four studies), viruses (seven studies), cells (three studies), and oligonucleotides (one study). Overall, all types of interventions increased flap survival, with treatment using anti-NF-κB oligonucleotide achieving the best results and treatment with KGF-containing virus, the lowest.

2.2. Secondary Outcome–Vessel Density

The authors measured vessel density using a variety of methods in 13 studies and 35 comparisons (Table S3) [7,11,12,13,15,20,33,41,42,45,47,49,53]. The overall effect estimate for all intervention types was an increase in standardized mean difference of 5.25 (95% CI 3.62–6.87), results were considerably heterogeneous (I2 = 93%), and intervention subgroups differed (p < 0.01) (Figure 7).
We performed a meta-regression considering administration time, assessment time, and type of intervention (Table S4) with a significant test of moderators (p = 0.03). Assessment time (SMD 1.0, 95% CI 0.38–1.63) and VEGF intervention (SMD 4.85, 95% CI 0.08–9.62) were significant positive moderators of the effect size. The model accounted for 9.10% of total heterogeneity. There was a significant publication bias reflected in a funnel plot asymmetry and a significant (p < 0.001) Egger’s test result.

2.3. Risk of Bias

Studies were assessed using the SYRCLE Risk of Bias Tool for Animal Studies. The average score was 4.34 out of 10 points (Table S5). The most frequently reported sources of potential bias included the lack of random housing of animals (n = 49), blinding following intervention (n = 49), outcome assessor blinding (n = 45), allocation concealment (n = 42), allocation sequence randomization (n = 37), and random animal selection for outcome measurements (n = 39). Few studies did not address incomplete outcome data (n = 7). Other sources of bias were also found in some of the studies (n = 11). This included imprecise data presentation, a low number of animals/flaps in the experimental groups, dubious lack of animal loss, uncommonly low SD values, and no clear definition of primary and secondary outcomes.

2.4. Certainty of Evidence

The assessment of certainty of evidence using GRADE has shown that only VEGF treatment may improve flap survival and vessel density (Tables S6–S10). For all other interventions, it is uncertain if they improve any outcomes as the certainty of evidence is very low.

3. Discussion

3.1. Main Findings

We found that treatment with locally administered growth factors delivered via vectors increased surgical flap survival in vivo by between 4% (FGF) and 16% (VEGF). The primary outcome was most assessed on day 7. The studied interventions also enhanced angiogenesis, increasing vessel density by an SMD of over 5, a time-dependent effect most prominently observed with VEGF. Although many studies were included in this meta-analysis, identifying the key factors influencing the outcome effect estimates is challenging. The levels of statistical heterogeneity were substantial or considerable for most analyses and could arise from differences in study design, systematic error (bias), applied interventions, and random error.
Subgroup analysis showed that VEGF administration had the highest effect estimate for flap survival improvement in muscular flaps (TRAM) among other flap types. However, this effect was the least precise. Thus, we can only hypothesize that the underlying mechanism may be related to poor ischemia tolerance in muscular flaps, resulting in greater improvements if treated [58].
Seemingly important study characteristics, such as the route of administration, the timing of the intervention, or the vectors used, failed to explain the differences in effect sizes between studies, as the meta-regression showed. Only 9% of total heterogeneity among studies using VEGF could be accounted for, taking the above-mentioned factors into consideration. Different surgical flaps were used across studies; however, in most cases, they showed unanimously beneficial effects of locally overproduced growth factors.
Despite inconclusive results from meta-regression concerning effect moderators, several studies have provided indicative data.
Delivery time is an important determinant for the further translation of the results. We lack high-quality studies with head-to-head comparisons of different preoperative administration times, as only two studies of acceptable quality addressed this issue [30,34]. Giunta et al. [30] indicated that increasing the delay between adenoviral administration and flap elevation by up to 7 days is beneficial. Chen et al. [34] used modified myoblasts and found that using intervention on preoperative day 4 resulted in better outcomes than on preoperative days 0, 2, or 7. They relied on same-animal internal controls, unlike most of the studies, which may explain the relatively narrow confidence intervals. As for the general characteristics of viral transduction [59], if viral or cellular delivery is planned, some delay in flap harvesting should be considered (4–7 days).
We lacked data on the optimal flap harvesting time following plasmid-based gene therapy. Based on the summarized results, we hypothesized that if the aim of the therapy was to prevent flap ischemia in the early postoperative period, the intraoperative use of a plasmid containing VEGF may be considered. However, further deductions are limited, as details concerning promoter and plasmid delivery form (e.g., liposomes, electroporation, etc.) were not considered in the meta-analysis. In both random and free flaps, direct tissue injections (subcutaneous, intradermal, and intramuscular) were used most frequently. Free flap or pedicled flap use enables consideration of intravascular administration of genetic therapy. Only two studies [38,51] used intra-arterial perfusion of flaps to administer either VEGF or anti-NF-κB oligonucleotides, both achieving high flap survival. However, single studies with a high risk of bias, using different interventions, are insufficient to confirm the superiority of this administration route. In addition, the advantages of intravascular therapy should be considered alongside potential risks (yet unreported) of thrombosis after injury to the pedicle, irritation of the intima with a high concentration of particles, and, by default, limited use of certain flap types.
The isoforms of target proteins should not be overlooked. Rinsch et al. [39] showed that VEGF-167 is significantly more efficient than the other forms of VEGF. This may be related to its ischemia-specific angiogenic activity previously observed in the myocardium and its role as a fatty acid metabolism regulator [60]. Compared with VEGF-165, which is the main angiogenesis regulator, VEGF-167 may be more specific to unstable conditions in the flap microenvironment.
Using multiple proteins does not seem to improve flap survival, as combined VEGF + FGF treatment was not superior to other interventions. This finding is limited by the small number of included studies. Although these cytokines are known to work synergistically [61], the lack of a clinically meaningful synergistic effect may be explained by the delivery mode resulting in metabolic overactivation following intense plasmid transfection.
Meta-regressions and sensitivity analyses did not prove that any of the parameters related to vectors—either cells, viruses, or plasmid produce a significant difference for the primary outcome. Thus, further research on pharmacodynamics and pharmacokinetics should be designed.
Interestingly, only Uemura et al. [51] reported the use of intra-arterially administered oligonucleotides, showing a gap in knowledge regarding this treatment modality. As this treatment provided the best results among the qualitatively described studies, further investigation based on this vehicle and the route of delivery is warranted [62].
Currently, there are 32 FDA-approved cellular or gene therapies, proving that contemporary techniques are sufficiently safe and efficient for clinical use [63]. The greatest controversies and concerns related to gene therapy are related to potential systemic cyto- and genotoxicity or unpredictable distant side effects in nontarget locations. However, none of the included studies reported such.

3.2. Limitations of the Study

Although a beneficial effect was clearly observed, the results showed substantial statistical heterogeneity and at least a considerable risk of bias. Such heterogeneity is not uncommon in preclinical studies and can be explained by the lack of unified housing conditions and the varying strains of animals used. It is noteworthy that studies were conducted over a relatively wide period of over 20 years, during which standards of conducting and reporting animal studies have altered significantly, especially in the European Union, due to novel regulations [64]. Studies included in this review were generally of poor quality as per the risk of bias assessment, mostly due to inappropriate randomization and blinding. Relatively low SDs may require further consideration of the quality and reliability of reported data. This phenomenon was explicitly observed in studies with a low animal/flap count. A high risk of bias in the selected studies, coupled with surprisingly narrow confidence intervals, is likely the main reason for the considerable statistical heterogeneity. They would also explain why the meta-regression model accounted for a small percentage of the total heterogeneity. As this was the first meta-analysis in the field, we did not want to restrict the inclusion criteria. Access to comprehensive data reports was limited, and most studies did not publish supplementary datasets. The results had to be extracted based on the values found in the main text or directly from the figures.
Overall, despite the beneficial effects of some molecules (particularly VEGF), the quality of evidence is low to very low, undermined mainly by a high risk of bias in the included studies and differing levels of random error.

4. Methods

This study was undertaken in accordance with the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines [7], and the study protocol was registered in the PROSPERO database (CRD42022S359982). PRISMA checklist is available in Supplementary Material (Table S1).

4.1. Search Strategy

A comprehensive and systematic search of PubMed, Embase, Web of Science, and Scopus was conducted on 11 December 2022 and updated on 8 January 2024 to identify relevant studies published until the date of the search. We combined controlled vocabulary (MeSH Terms, Emtree terms) with keywords to produce a search engine for each database. Terms related to surgical flaps and genetic modifications delivered via bacterial, viral, or cellular vectors were implemented. The following Medical Subject Headings were used: genetic therapy, cell transplantation, mesenchymal stem cells, bacteria, and drug carriers. The full search strategy is available in the PROSPERO report.

4.2. Inclusion Criteria

The following studies were included: studies that compared at least one control group with a treatment group, studies with different animal models, studies in which genetic modification of a tissue bulk of a living organism or genetic modification of cells/tissues later integrated with the former tissue bulk were used in the treatment group, and studies in which the control group received sham genetic modification or were treated using previously established standards.

4.3. Exclusion Criteria

Studies excluded were human studies, only in vitro studies that included split-thickness skin grafts, studies in which flaps received additional ischemia after the intervention, studies that lacked any intervention in the control group, studies reported in languages other than English, studies that did not report any unified measurement of flap survival, and studies with severe flaws in the reported data.

4.4. Primary Outcome

The primary outcome was the percentage of flap survival after n days post-intervention or the percentage of necrotic tissue within the flap. Data reported as a percentage of flap necrosis were later transformed into a percentage of flap survival to unify the overall outcome.

4.5. Secondary Outcomes

The secondary outcomes included the median number of vessels in the high-power field, mean number of vessels in the high-power field, number of capillaries per mm2, and improvement of other parameters treated with the modified flap. The results reported in values other than the median number of vessels in the high-power field were transformed (where applicable) into such data.
Titles and abstracts of studies retrieved using the search strategy were screened using the Rayyan QCRI to identify studies that met the inclusion criteria. All screenings were independently performed by two review team members (M.G., M.K.). Any disagreement regarding the eligibility of the studies was resolved through discussion with a third author when necessary (W.P.).

4.6. Data Extraction, Collection and Synthesis

All the data were extracted using a predefined extraction form. The following information was collected: authors, year, information about the animal model (species, strain, sex, weight), delivered drug (name, mode, and route of delivery, dose), and treatment (time of administration and assessment, flap type, and size). For continuous outcomes, the mean, standard deviation, and sample size for each study group were obtained. When necessary, data were extracted from the figures using WebPlotDigitizer v.4.6. If only standard errors were provided, they were transformed into standard deviations, multiplied by the square root of the sample size. To avoid unit of analysis issues, for studies with more than one intervention group and only one control group, we divided the number of animals in the control group by the number of intervention groups or combined data from the intervention groups if they were sufficiently similar. All changes are described in Supplementary Material (List S1).

4.7. Statistical Analysis

All analyses were performed using R version 4.3.1 and RStudio. Results for the primary outcome (flap survival) were presented as mean differences with 95% confidence intervals. Statistical heterogeneity was determined using the I2 statistic. For all outcomes, we used the DerSimonian–Laird random-effects model meta-analysis because we anticipated significant heterogeneity in the administration and assessment times as well as dosages, vectors, and flap types. As planned, we performed separate meta-analyses for each identified intervention and subgroup analysis for each flap and vector type. We performed a quantitative synthesis only if at least three comparisons reporting the outcomes of a given intervention were available. To ascertain the robustness of the findings, we performed three sensitivity analyses: (1) excluding studies assessed to be at a high risk of bias (i.e., scored less than 4 points on the SYRCLE checklist), (2) excluding data extracted from figures and not reported explicitly in tables or manuscripts, and (3) using an inverse-variance fixed-effect model rather than a random-effects model. A separate sensitivity analysis was planned based on the time between drug administration and flap elevation. Due to a variety of reported time gaps and a multitude of identified studies, we decided to perform a meta-regression for all studies evaluating the efficacy of VEGF. Other study characteristics were also included in this model, including administration time, assessment time, vector type, and route of administration. Three individual studies that reported rare routes of administration (intra-arterial, intrafascial, and topical) were excluded from the model to avoid generating spurious associations. Results are presented as effect estimates with 95% confidence intervals. For the subgroup analyses, we tested the interactions using the χ2 significance test.
The secondary outcome (blood vessel density) was reported using different tools and measures in the included studies. Therefore, results in this case are presented as standardized mean differences (SMDs) using Hedge’s h with corresponding 95% CIs. We also performed a meta-regression to analyze how an overall effect estimate was influenced by potential moderators: administration time, assessment time, and intervention type. Results are presented as effect estimates with 95% confidence intervals.
Publication bias was assessed for analyses including at least ten comparisons, funnel plots were generated and visually inspected, and a formal Egger’s test was performed.

4.8. Risk of Bias (Quality) Assessment

The quality of the included studies was determined using the SYRCLE checklist for study quality [65]. The two reviewers (M.G., W.P.) were blinded, and the quality of the studies was assessed separately. Discrepancies were discussed with a third reviewer to reach a consensus (A.S.).

4.9. Certainty of Evidence Assessment

The certainty of evidence was determined using the GRADE approach [66], modified for use in the preclinical meta-analysis [67]. M.G. and A.S. assessed the certainty of evidence independently. Any discrepancies and clinical relevance were consulted with the third reviewer (W.P.) to reach a consensus.

5. Conclusions

  • VEGF gene delivery to the flap increases its survival regardless of flap type, gene method, or timing in an in vivo model.
  • Other proteins, such as FGF, HGF, and PDGF, may also be beneficial, but to a lesser extent.
  • The quality of animal studies was predominantly low, which likely led to a high level of unexplained heterogeneity; however, general quantitative analysis is feasible.
  • Translational studies appear to be sufficiently founded, yet any new interventions should be preceded by high-quality preclinical analysis.
  • Additional, targeted, and detailed reviews and original studies should be performed to summarize the knowledge of pharmacodynamics and pharmacokinetics of gene therapies in flaps.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms25052622/s1.

Author Contributions

Conceptualization, W.P. and P.W.; methodology, A.S.; software, A.S.; validation, W.P., M.G. and A.S.; formal analysis, W.P., M.G., A.S. and M.K.; investigation, W.P., M.G., A.S. and M.K.; resources, W.P. and P.W.; data curation, W.P., A.S. and P.W.; writing—original draft preparation, W.P., M.G., A.S. and M.K.; writing—review and editing, W.P., M.G. and A.S.; visualization, A.S.; supervision, P.W.; project administration, W.P. and P.W.; funding acquisition, W.P. and P.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ministry of Science and Higher Education of Poland, grant no: 0075/DIA/2017/46. Recipient: WP.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Moellhoff, N.; Taha, S.; Wachtel, N.; Hirschmann, M.; Hellweg, M.; Giunta, R.E.; Ehrl, D. Analysis of Factors Determining Patient Survival after Receiving Free-Flap Reconstruction at a Single Center-A Retrospective Cohort Study. Diagnostics 2022, 12, 2877. [Google Scholar] [CrossRef]
  2. Harder, Y.; Amon, M.; Laschke, M.W.; Schramm, R.; Rücker, M.; Wettstein, R.; Bastiaanse, J.; Frick, A.; Machens, H.G.; Küntscher, M.; et al. An old dream revitalised: Preconditioning strategies to protect surgical flaps from critical ischaemia and ischaemia-reperfusion injury. J. Plast. Reconstr. Aesthet. Surg. 2008, 61, 503–511. [Google Scholar] [CrossRef]
  3. Lee, J.-H.; You, H.-J.; Lee, T.-Y.; Kang, H.J. Current Status of Experimental Animal Skin Flap Models: Ischemic Preconditioning and Molecular Factors. Int. J. Mol. Sci. 2022, 23, 5234. [Google Scholar] [CrossRef] [PubMed]
  4. Afrooghe, A.; Damavandi, A.R.; Ahmadi, E.; Jafari, R.M.; Dehpour, A.R. The current state of knowledge on how to improve skin flap survival: A review. J. Plast. Reconstr. Aesthet. Surg. 2023, 82, 48–57. [Google Scholar] [CrossRef]
  5. Wolff, J.A.; Malone, R.W.; Williams, P.; Chong, W.; Acsadi, G.; Jani, A.; Felgner, P.L. Direct Gene Transfer into Mouse Muscle in Vivo. Science 1990, 247, 1465–1468. [Google Scholar] [CrossRef] [PubMed]
  6. Takeshita, S.; Weir, L.; Chen, D.; Zheng, L.P.; Riessen, R.; Bauters, C.; Symes, J.F.; Ferrara, N.; Isner, J.M. Therapeutic angiogenesis following arterial gene transfer of vascular endothelial growth factor in a rabbit model of hindlimb ischemia. Biochem. Biophys. Res. Commun. 1996, 227, 628–635. [Google Scholar] [CrossRef]
  7. Neumeister, M.W.; Song, Y.H.; Mowlavi, A.; Suchy, H.; Mathur, A. Effects of liposome-mediated gene transfer of VEGF in ischemic rat gracilis muscle. Microsurgery 2001, 21, 58–62. [Google Scholar] [CrossRef] [PubMed]
  8. Michlits, W.; Mittermayr, R.; Schafer, R.; Redl, H.; Aharinejad, S. Fibrin-embedded administration of VEGF plasmid enhances skin flap survival. Wound Repair Regen. 2007, 15, 360–367. [Google Scholar] [CrossRef]
  9. Meirer, R.; Huemer, G.M.; Oehlbauer, M.; Wanner, S.; Piza-Katzer, H.; Kamelger, F.S. Comparison of the effectiveness of gene therapy with vascular endothelial growth factor or shock wave therapy to reduce ischaemic necrosis in an epigastric skin flap model in rats. J. Plast. Reconstr. Aesthet. Surg. 2007, 60, 266–271. [Google Scholar] [CrossRef]
  10. Lubiatowski, P.; Goldman, C.K.; Gurunluoglu, R.; Carnevale, K.; Siemionow, M. Enhancement of epigastric skin flap survival by adenovirus-mediated VEGF gene therapy. Plast. Reconst. Surg. 2002, 109, 1986–1993. [Google Scholar] [CrossRef]
  11. Liu, P.Y.; Wang, X.T.; Badiavas, E.; Rieger-Christ, K.; Tang, J.B.; Summerhayes, I. Enhancement of ischemic flap survival by prefabrication with transfer of exogenous PDGF gene. J. Reconstr. Microsurg. 2005, 21, 273–279. [Google Scholar] [CrossRef]
  12. Liu, P.Y.; Tong, W.; Liu, K.; Han, S.H.; Wang, X.T.; Badiavas, E.; Rieger-Christ, K.; Summerhayes, I. Liposome-mediated transfer of vascular endothelial growth factor cDNA augments survival of random-pattern skin flaps in the rat. Wound Repair Regen. 2004, 12, 80–85. [Google Scholar] [CrossRef]
  13. Liu, P.Y.; Liu, K.; Wang, X.T.; Badiavas, E.; Rieger-Christ, K.M.; Tang, J.B.; Summerhayes, I.C. Efficacy of combination gene therapy with multiple growth factor cDNAs to enhance skin flap survival in a rat model. DNA Cell Biol. 2005, 24, 751–757. [Google Scholar] [CrossRef] [PubMed]
  14. Lasso, J.M.; Del Río, M.; García, M.; Martínez Calleja, V.; Nava, P.; Muñoz-Fernández, M.A.; Pérez Cano, R. Improving flap survival by transplantation of a VEGF-secreting endothelised scaffold during distal pedicle flap creation. J. Plast. Reconstr. Aesthet. Surg. 2007, 60, 279–286. [Google Scholar] [CrossRef]
  15. Huang, N.; Khan, A.; Ashrafpour, H.; Neligan, P.C.; Forrest, C.R.; Kontos, C.D.; Pang, C.Y. Efficacy and mechanism of adenovirus-mediated VEGF-165 gene therapy for augmentation of skin flap viability. Am. J. Physiol. Heart Circ. 2006, 291, H127–H137. [Google Scholar] [CrossRef] [PubMed]
  16. Holzbach, T.; Vlaskou, D.; Neshkova, I.; Konerding, M.A.; Wörtler, K.; Mykhaylyk, O.; Gänsbacher, B.; Machens, H.G.; Plank, C.; Giunta, R.E. Non-viral VEGF165 gene therapy-magnetofection of acoustically active magnetic lipospheres (‘magnetobubbles’) increases tissue survival in an oversized skin flap model. J. Cell. Mol. Med. 2010, 14, 587–599. [Google Scholar] [CrossRef]
  17. Hijjawi, J.; Mogford, J.E.; Chandler, L.A.; Cross, K.J.; Said, H.; Sosnowski, B.A.; Mustoe, T.A. Platelet-derived growth factor B, but not fibroblast growth factor 2, plasmid DNA improves survival of ischemic myocutaneous flaps. Arch. Surg. 2004, 139, 142–147. [Google Scholar] [CrossRef] [PubMed]
  18. Machens, H.G.; Morgan, J.R.; Berthiaume, F.; Stefanovich, P.; Siemers, F.; Krapohl, B.; Berger, A.; Mailänder, P. Platelet-derived growth factor-AA-mediated functional angiogenesis in the rat epigastric island flap after genetic modification of fibroblasts is ischemia dependent. Surgery 2002, 131, 393–400. [Google Scholar] [CrossRef] [PubMed]
  19. Nakagawa, A.; Makino, H.; Aoki, M.; Miyake, T.; Shiraya, S.; Nakamura, T.; Ogihara, T.; Kimata, Y.; Morishita, R. Improvement of survival of skin flaps by combined gene transfer of hepatocyte growth factor and prostacyclin synthase. J. Gene Med. 2007, 9, 1087–1094. [Google Scholar] [CrossRef]
  20. Rah, D.K.; Yun, I.S.; Yun, C.O.; Lee, S.B.; Lee, W.J. Gene therapy using hepatocyte growth factor expressing adenovirus improves skin flap survival in a rat model. J. Korean Med. Sci. 2014, 29 (Suppl. 3), S228–S236. [Google Scholar] [CrossRef]
  21. Huemer, G.M.; Shafighi, M.; Meirer, R.; Debagge, P.; Piza-Katzer, H.; Gurunluoglu, R. Adenovirus-mediated transforming growth factor-β ameliorates ischemic necrosis of epigastric skin flaps in a rat model. J. Surg. Res. 2004, 121, 101–107. [Google Scholar] [CrossRef]
  22. Huemer, G.M.; Meirer, R.; Gurunluoglu, R.; Kamelger, F.S.; Dunst, K.M.; Wanner, S.; Piza-Katzer, H. Comparison of the effectiveness of gene therapy with transforming growth factor-β or extracorporal shock wave therapy to reduce ischemic necrosis in an epigastric skin flap model in rats. Wound Repair Regen. 2005, 13, 262–268. [Google Scholar] [CrossRef]
  23. Liu, P.Y.; Wang, X.T.; Xin, K.Q.; Chen, C.H.; Rieger-Christ, K.; Summerhayes, I.C.; Fang Wu, Y.; Tang, J.B. Application of AAV2-mediated bFGF gene therapy on survival of ischemic flaps: Effects of timing of gene transfer. Ann. Plast. Surg. 2009, 62, 87–91. [Google Scholar] [CrossRef]
  24. Lee, W.J.; Yun, C.O.; Yun, I.S.; Kim, Y.O.; Choi, I.K.; Yun, T.J.; Rah, D.K. Augmentation of rat skin flap viability by relaxin-expressing adenovirus. Wound Repair Regen. 2011, 19, 709–717. [Google Scholar] [CrossRef]
  25. Jung, H.; Gurunluoglu, R.; Scharpf, J.; Siemionow, M. Adenovirus-mediated angiopoietin-1 gene therapy enhances skin flap survival. Microsurgery 2003, 23, 374–380. [Google Scholar] [CrossRef]
  26. Choi, S.W.; Jeon, Y.R.; Baek, W.; Yun, C.O.; Roh, T.S.; Lee, W.J. Dickkopf 2-Expressing Adenovirus Increases the Survival of Random-Pattern Flaps and Promotes Vasculogenesis in a Rat Model. Ann. Plast. Surg. 2020, 84, 588–594. [Google Scholar] [CrossRef]
  27. Lou, J.; Wang, X.; Zhang, H.; Yu, G.; Ding, J.; Zhu, X.; Li, Y.; Wu, Y.; Xu, H.; Gao, W.; et al. Inhibition of PLA2G4E/cPLA2 promotes survival of random skin flaps by alleviating Lysosomal membrane permeabilization-Induced necroptosis. Autophagy 2021, 18, 1841–1863. [Google Scholar] [CrossRef] [PubMed]
  28. Gurunluoglu, R.; Ozer, K.; Skugor, B.; Lubiatowski, P.; Carnevale, K.; Siemionow, M. Effect of transfection time on the survival of epigastric skin flaps pretreated with adenovirus encoding the VEGF gene. Ann. Plast. Surg. 2002, 49, 161–169. [Google Scholar] [CrossRef] [PubMed]
  29. Gurunluoglu, R.; Meirer, R.; Shafighi, M.; Huemer, G.M.; Yilmaz, B.; Piza-Katzer, H. Gene therapy with adenovirus-mediated VEGF enhances skin flap prefabrication. Microsurgery 2005, 25, 433–441. [Google Scholar] [CrossRef] [PubMed]
  30. Giunta, R.E.; Holzbach, T.; Taskov, C.; Holm, P.S.; Konerding, M.A.; Schams, D.; Biemer, E.; Gänsbacher, B. AdVEGF165 gene transfer increases surviral in overdimensioned skin flaps. J. Gene Med. 2005, 7, 297–306. [Google Scholar] [CrossRef] [PubMed]
  31. Fujihara, Y.; Koyama, H.; Nishiyama, N.; Eguchi, T.; Takato, T. Gene transfer of bFGF to recipient bed improves survival of ischemic skin flap. Br. J. Surg. 2005, 58, 511–517. [Google Scholar] [CrossRef] [PubMed]
  32. Ferraro, B.; Cruz, Y.L.; Coppola, D.; Heller, R. Intradermal delivery of plasmid VEGF-165 by electroporation promotes wound healing. Mol. Ther. 2009, 17, 651–657. [Google Scholar] [CrossRef] [PubMed]
  33. de Freitas, A.L.P.; Gomes, H.C.; Lisboa, B.C.G.; Arias, V.; Han, S.W.; Ferreira, L.M. Effect of gene therapy with vascular endothelial growth factor after abdominoplasty on TRAM flap viability in a rat model. Plast. Reconst. Surg. 2010, 125, 1343–1351. [Google Scholar] [CrossRef] [PubMed]
  34. Chen, W.; Yang, D.; Wang, P.; Gao, S.; Zhang, X.; Wang, T. Microencapsulated myoblasts transduced by the vascular endothelial growth factor (VEGF) gene for the ischemic skin flap. Aesthetic Plast. Surg. 2011, 35, 326–332. [Google Scholar] [CrossRef] [PubMed]
  35. Chang, K.H.; Shoureshi, P.; Lay, F.; Sebastian, R.; Alikhassy Habibabady, Z.; Born, L.J.; Marti, G.P.; Meltzer, S.J.; Abraham, J.M.; Harmon, J.W. Preconditioning of surgical pedicle flaps with DNA plasmid expressing hypoxia-inducible factor-1α (HIF-1α) promotes tissue viability. Gene Ther. 2021, 28, 319–328. [Google Scholar] [CrossRef] [PubMed]
  36. Basu, G.; Downey, H.; Guo, S.; Israel, A.; Asmar, A.; Hargrave, B.; Heller, R. Prevention of distal flap necrosis in a rat random skin flap model by gene electrotransfer delivering VEGF-165 plasmid. J. Gene Med. 2014, 16, 55–65. [Google Scholar] [CrossRef] [PubMed]
  37. Antonini, A.; Zacchigna, S.; Papa, G.; Novati, F.; Pascone, M.; Giacca, M. Improved survival of rat ischemic cutaneous and musculocutaneous flaps after VEGF gene transfer. Microsurgery 2007, 27, 439–445. [Google Scholar] [CrossRef] [PubMed]
  38. Taub, P.J.; Marmur, J.D.; Zhang, W.X.; Senderoff, D.; Urken, M.L.; Silver, L.; Weinberg, H. Effect of time on the viability of ischemic skin flaps treated with vascular endothelial growth factor (VEGF) cDNA. J. Reconstr. Microsurg. 1998, 14, 387–390. [Google Scholar] [CrossRef]
  39. Rinsch, C.; Quinodoz, P.; Pittet, B.; Alizadeh, N.; Baetens, D.; Montandon, D.; Aebischer, P.; Pepper, M.S. Delivery of FGF-2 but not VEGF by encapsulated genetically engineered myoblasts improves survival and vascularization in a model of acute skin flap ischemia. Gene Ther. 2001, 8, 523–533. [Google Scholar] [CrossRef]
  40. O’Toole, G.; MacKenzie, D.; Lindeman, R.; Buckley, M.F.; Marucci, D.; McCarthy, N.; Poole, M. Vascular endothelial growth factor gene therapy in ischaemic rat skin flaps. Br. J. Plast. Surg. 2002, 55, 55–58. [Google Scholar] [CrossRef]
  41. Yang, L.W.; Zhang, J.X.; Zeng, L.; Xu, J.J.; Du, F.T.; Luo, W.; Luo, Z.J.; Jiang, J.H. Vascular endothelial growth factor gene therapy with intramuscular injections of plasmid DNA enhances the survival of random pattern flaps in a rat model. Br. J. Plast. Surg. 2005, 58, 339–347. [Google Scholar] [CrossRef] [PubMed]
  42. Zacchigna, S.; Papa, G.; Antonini, A.; Novati, F.; Moimas, S.; Carrer, A.; Arsic, N.; Zentilin, L.; Visintini, V.; Pascone, M.; et al. Improved survival of ischemic cutaneous and musculocutaneous flaps after vascular endothelial growth factor gene transfer using adeno-associated virus vectors. Am. J. Pathol. 2005, 167, 981–991. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, F.; Yang, F.; Hu, E.C.; Sones, W.; Lei, M.; Lineaweaver, W.C. Vascular endothelial growth factor gene therapy in improvement of skin paddle survival in a rat TRAM flap model. J. Reconstr. Microsurg. 2005, 21, 391–396. [Google Scholar] [CrossRef]
  44. Wang, X.T.; Liu, P.Y.; Tang, J.B. PDGF gene therapy enhances expression of VEGF and bFGF genes and activates the NF-κB gene in signal pathways in ischemic flaps. Plast. Reconst. Surg. 2006, 117, 129–137. [Google Scholar] [CrossRef]
  45. Yi, C.; Xia, W.; Zheng, Y.; Zhang, L.; Shu, M.; Liang, J.; Han, Y.; Guo, S. Transplantation of endothelial progenitor cells transferred by vascular endothelial growth factor gene for vascular regeneration of ischemic flaps. J. Surg. Res. 2006, 135, 100–106. [Google Scholar] [CrossRef] [PubMed]
  46. Zheng, Y.; Yi, C.; Xia, W.; Ding, T.; Zhou, Z.; Han, Y.; Guo, S. Mesenchymal stem cells transduced by vascular endothelial growth factor gene for ischemic random skin flaps. Plast. Reconst. Surg. 2008, 121, 59–69. [Google Scholar] [CrossRef]
  47. Spanholtz, T.; Maichle, A.; Niedworok, C.; Stoeckelhuber, B.M.; Krüger, S.; Wedel, T.; Aach, T.; Middeler, G.; Hellwig-Bürgel, T.; Bader, A.; et al. Timing and Targeting of Cell-Based VEGF165 Gene Expression in Ischemic Tissue. J. Surg. Res. 2009, 151, 153–162. [Google Scholar] [CrossRef]
  48. Rezende, F.C.; Gomes, H.C.; Lisboa, B.; Lucca, A.F.; Han, S.W.; Ferreira, L.M. Electroporation of vascular endothelial growth factor gene in a unipedicle transverse rectus abdominis myocutaneous flap reduces necrosis. Ann. Plast. Surg. 2010, 64, 242–246. [Google Scholar] [CrossRef]
  49. Spanholtz, T.A.; Theodorou, P.; Holzbach, T.; Wutzler, S.; Giunta, R.E.; MacHens, H.G. Vascular endothelial growth factor (VEGF 165) plus basic fibroblast growth factor (bFGF) producing cells induce a mature and stable vascular network-A future therapy for ischemically challenged tissue. J. Surg. Res. 2011, 171, 329–338. [Google Scholar] [CrossRef]
  50. Wang, X.T.; Avanessian, B.; Ma, Q.; Durfee, H.; Tang, Y.Q.; Liu, P.Y. Enhancement of flap survival and changes in angiogenic gene expression after AAV2-mediated VEGF gene transfer to rat ischemic flaps. Wound Repair Regen. 2011, 19, 498–504. [Google Scholar] [CrossRef]
  51. Uemura, T.; Tsujii, M.; Akeda, K.; Iino, T.; Satonaka, H.; Hasegawa, M.; Sudo, A. Transfection of nuclear factor-kappaB decoy oligodeoxynucleotide protects against ischemia/reperfusion injury in a rat epigastric flap model. J. Gene Med. 2012, 14, 623–631. [Google Scholar] [CrossRef]
  52. Wang, X.; Yu, M.; Zhu, W.; Bao, T.; Zhu, L.; Zhao, W.; Zhao, F.; Wang, H. Adenovirus-mediated expression of keratinocyte growth factor promotes secondary flap necrotic wound healing in an extended animal model. Aesthetic Plast. Surg. 2013, 37, 1023–1033. [Google Scholar] [CrossRef] [PubMed]
  53. Seyed Jafari, S.M.; Shafighi, M.; Beltraminelli, H.; Geiser, T.; Hunger, R.E.; Gazdhar, A. Improvement of Flap Necrosis in a Rat Random Skin Flap Model by In Vivo Electroporation-Mediated HGF Gene Transfer. Plast. Reconst. Surg. 2017, 139, 1116e–1127e. [Google Scholar] [CrossRef] [PubMed]
  54. Seyed Jafari, S.M.; Shafighi, M.; Beltraminelli, H.; Weber, B.; Schmid, R.A.; Geiser, T.; Gazdhar, A.; Hunger, R.E. Efficacy of In Vivo Electroporation-Mediated IL-10 Gene Delivery on Survival of Skin Flaps. J. Membr. Biol. 2018, 251, 211–219. [Google Scholar] [CrossRef] [PubMed]
  55. Seyed Jafari, S.M.; Blank, F.; Ramser, H.E.; Woessner, A.E.; Shafighi, M.; Geiser, T.; Quinn, K.P.; Hunger, R.E.; Gazdhar, A. Efficacy of Combined in-vivo Electroporation-Mediated Gene Transfer of VEGF, HGF, and IL-10 on Skin Flap Survival, Monitored by Label-Free Optical Imaging: A Feasibility Study. Front. Surg. 2021, 8, 639661. [Google Scholar] [CrossRef] [PubMed]
  56. Zhang, F.-G.; Yao, Y.; Feng, Y.; Hua, C.-G.; Tang, X.-F. Mesenchymal Stem Cells Transduced by Stromal Cell–Derived Factor-1α Augment Ischemic Free Flaps’ Survival. Ann. Plast. Surg. 2011, 66, 92–97. [Google Scholar] [CrossRef] [PubMed]
  57. Luo, Z.; Bian, Y.; Zheng, G.; Wang, H.; Yan, B.; Su, W.; Dong, W.; Hu, Z.; Ding, J.; Wang, A.; et al. Chemically Modified SDF-1α mRNA Promotes Random Flap Survival by Activating the SDF-1α/CXCR4 Axis in Rats. Front. Cell Dev. Biol. 2021, 9, 623959. [Google Scholar] [CrossRef] [PubMed]
  58. Lee, Z.H.; Abdou, S.A.; Daar, D.A.; Anzai, L.; Stranix, J.T.; Thanik, V.; Levine, J.P.; Saadeh, P.B. Comparing Outcomes for Fasciocutaneous versus Muscle Flaps in Foot and Ankle Free Flap Reconstruction. J. Reconstr. Microsurg. 2019, 35, 646–651. [Google Scholar] [CrossRef]
  59. Sara, C.; David, M.; Simon, R.; Garrett, C.; Martina, S.; Patrick, H.; Gerald, O.S.; Mark, T. Comparison of DNA Delivery and Expression Using Frequently Used Delivery Methods. In Gene Ther.; IntechOpen: Rijeka, Croatia, 2011. [Google Scholar]
  60. Li, X. VEGF-B: A thing of beauty. Cell Res. 2010, 20, 741–744. [Google Scholar] [CrossRef]
  61. Murakami, M.; Nguyen, L.T.; Hatanaka, K.; Schachterle, W.; Chen, P.-Y.; Zhuang, Z.W.; Black, B.L.; Simons, M. FGF-dependent regulation of VEGF receptor 2 expression in mice. J. Clin. Investig. 2011, 121, 2668–2678. [Google Scholar] [CrossRef]
  62. Roberts, T.C.; Langer, R.; Wood, M.J.A. Advances in oligonucleotide drug delivery. Nat. Rev. Drug Discov. 2020, 19, 673–694. [Google Scholar] [CrossRef] [PubMed]
  63. Approved Cellular and Gene Therapy Products. Available online: https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/approved-cellular-and-gene-therapy-products (accessed on 27 August 2023).
  64. Directive 2010/63/EU of the European Parliament and of the Council of 22 September 2010 on the protection of animals used for scientific purposes Text with EEA relevance. Off. J. Eur. Union 2010, L 276, 33–79.
  65. Hooijmans, C.R.; Rovers, M.M.; de Vries, R.B.M.; Leenaars, M.; Ritskes-Hoitinga, M.; Langendam, M.W. SYRCLE’s risk of bias tool for animal studies. BMC Med. Res. Methodol. 2014, 14, 43. [Google Scholar] [CrossRef] [PubMed]
  66. Granholm, A.; Alhazzani, W.; Møller, M.H. Use of the GRADE approach in systematic reviews and guidelines. Br. J. Anaesth. 2019, 123, 554–559. [Google Scholar] [CrossRef] [PubMed]
  67. Hooijmans, C.R.; de Vries, R.B.M.; Ritskes-Hoitinga, M.; Rovers, M.M.; Leeflang, M.M.; IntHout, J.; Wever, K.E.; Hooft, L.; de Beer, H.; Kuijpers, T.; et al. Facilitating healthcare decisions by assessing the certainty in the evidence from preclinical animal studies. PLoS ONE 2018, 13, e0187271. [Google Scholar] [CrossRef]
Figure 1. PRISMA Flowchart.
Figure 1. PRISMA Flowchart.
Ijms 25 02622 g001
Figure 2. Mean difference in flap survival between VEGF treatment and control of included comparisons [7,8,9,10,11,12,13,15,16,28,29,30,32,33,34,37,38,39,40,41,42,43,45,46,47,48,50] with confidence intervals and effect estimates.
Figure 2. Mean difference in flap survival between VEGF treatment and control of included comparisons [7,8,9,10,11,12,13,15,16,28,29,30,32,33,34,37,38,39,40,41,42,43,45,46,47,48,50] with confidence intervals and effect estimates.
Ijms 25 02622 g002
Figure 3. Mean difference in flap survival between FGF treatment and control of included comparisons [13,17,23,31,39,49] with confidence intervals and effect estimates.
Figure 3. Mean difference in flap survival between FGF treatment and control of included comparisons [13,17,23,31,39,49] with confidence intervals and effect estimates.
Ijms 25 02622 g003
Figure 4. Mean difference in flap survival between PDGF treatment and control of included comparisons [11,17,18,44] with confidence intervals and effect estimates.
Figure 4. Mean difference in flap survival between PDGF treatment and control of included comparisons [11,17,18,44] with confidence intervals and effect estimates.
Ijms 25 02622 g004
Figure 5. Mean difference in flap survival between HGF or HGF + PGIS treatment and control of included comparisons [19,20,53] with confidence intervals and effect estimates.
Figure 5. Mean difference in flap survival between HGF or HGF + PGIS treatment and control of included comparisons [19,20,53] with confidence intervals and effect estimates.
Ijms 25 02622 g005
Figure 6. Mean difference in flap survival between VEGF + FGF treatment and control of included comparisons [13,49] with confidence intervals and effect estimates.
Figure 6. Mean difference in flap survival between VEGF + FGF treatment and control of included comparisons [13,49] with confidence intervals and effect estimates.
Ijms 25 02622 g006
Figure 7. Standardized mean difference in vessel density between all interventions and control of included comparisons [7,11,12,13,15,20,33,41,42,45,47,49,53] with confidence intervals and effect estimates.
Figure 7. Standardized mean difference in vessel density between all interventions and control of included comparisons [7,11,12,13,15,20,33,41,42,45,47,49,53] with confidence intervals and effect estimates.
Ijms 25 02622 g007
Table 1. Overview of studies included in primary outcome analysis. *—mode of expression based on gene expression assessment methods. **—if no standard deviation presented, results estimated based on graphical data available and not used for quantitative analysis.
Table 1. Overview of studies included in primary outcome analysis. *—mode of expression based on gene expression assessment methods. **—if no standard deviation presented, results estimated based on graphical data available and not used for quantitative analysis.
First Author; Year (*)—Included In Meta-AnalysisTargetVectorMode of Expression *Vector QuantityType of Control GroupFollow-UpFlap TypeAnimal CharacteristicsSurgical TechniqueControl Group Flap Survival (%, SD) **Treatment Group Flap Survival (%, SD) **
Neumeister; 2001 (*) [7]VEGF-165PlasmidUnspecified50 μLLipofectamine only7 daysMuscle12 Wistar male rats (300–325 g)Intramuscular injection after 4-h ischemia21.5 ± 2.048.8 ± 2.1
Michlits; 2007 (*) [8]VEGF-165PlasmidUnspecified100 μgNo treatment (A)
Fibrin-sealant (B)
Fibrin-sealant and sham plasmid (C)
7 daysEpigastric48 Sprague–Dawley male rats (300–350 g)Topical administration after induced ischemia (not specified)A—60.76 ± 10.20
B—74.30 ± 18.15
C—75.41 ± 19.19
86.54 ± 10.71
Meirer; 2007 (*) [9]VEGF (I) Shockwave (II)VirusUnspecified108 PFUNo treatment7 daysEpigastric30 Sprague–Dawley male rats (300–500 g)Subdermal injection82.67 ± 8.15I—90.42 ± 3.19
II—97.77 ± 4.34
Lubiatowski; 2002 (*) [10]VEGFVirusUnspecified108 PFUSham virus (A)
No treatment (B)
14 daysEpigastric30 Sprague–Dawley male rats (300–350 g)Subdermal injection two days before elevationA—82.15 ± 9.7
B—79.9 ± 3.3
97.55 ± 1.84
Liu; 2005 (*) [11]PDGF-B (I) VEGF (II)PlasmidUnspecified50 μgSaline (A)
Sham plasmid (B)
7 daysMcFarlane45 Sprague–Dawley female rats (250–300 g)Intradermal injection seven days before elevationA—52.3 ± 5.0
B—50.7 ± 5.9
I—60.8 ± 7.8
II—57.6 ± 5.2
Liu; 2004 (*) [12]VEGF-165PlasmidUnspecified50 μgSaline (A)
Sham plasmid (B)
7 daysMcFarlane32 Sprague–Dawley female rats (250 g)Intradermal injection seven days before elevationA—51.3 ± 2.45
B—51.1 ± 3.05
63.71 ± 3.2
Liu; 2005 (*) [13]VEGF-165 (I) FGF-β (II) PDGF-B (III)PlasmidUnspecified50 μgSham plasmid7 daysMcFarlane60 Sprague–Dawley female rats (250–300 g)Intradermal injection 7 days before flap elevation52.3 ± 5.0I—57.6 ± 5.2
II—55.3 ± 6.1
I + II—64.3 ± 8.7
I + III—57.6 ± 9.5
I + II + III—44.9 ± 2.7
Lasso; 2007 [14]VEGF-165Cells (transduction)Unspecified5 × 105 cellsNon-transduced cells6 daysAxial ear32 New Zealand white male rabbits (2500–3000 g)Topical administration onto the scaffold with pedicle divided after five or two days (1, 2)1—2.50 ± 7.07
2—51.25 ± 45.88
1—55.62 ± 38.95
2—95.62 ± 4.95
Huang; 2006 (*) [15]VEGF-165 (I) eNOS (II)VirusUnspecified5 × 108 PFUPBS (A)
Sham virus (B)
7 daysMcFarlane24 Sprague–Dawley male rats (350–375 g)Subdermal injection seven days before flap elevationA—56.0 ± 3.0
B—54.0 ± 2.0
I—67.0 ± 1.0
II—70.0 ± 3.0
Holzbach; 2010 (*) [16]VEGF-165Plasmid (magnetized)Unspecified200 µlSham plasmid7 daysMcFarlane46 Sprague–Dawley male rats (350 g)Subcutaneous injection with additional ultrasounds seven days before flap elevation49.2 ± 0.0874.3 ± 0.05
Hijjawi; 2004 (*) [17]PDGF-B (I)
FGF-β (II)
Plasmid (collagen matrix)Unspecified4.8 mgNo treatment7 daysMuscle24 Sprague–Dawley male rats (250–300 g)Subcutaneous injection seven days before flap elevation33.2 ± 1.3I—56.1 ± 1.25
II—32.7 ± 1.44
Machens; 2002 (*) [18]PDGF-ACells (transduction)Constant (up to 96 h)1 × 107 cellsSaline (A)
Cell medium (B)
Non-modified cells (C)
0 or 7 days (1,2)Epigastric80 Lewis inbred female rats (200–225 g)Intramuscular at the time of flap elevationA1—50.0 ± 9.0
A2—56.0± 8.0
B1—49.0 ± 11.0
B2—52.0 ± 10.0
C1—54.0 ± 9.0
C2—57.0 ± 7.0
1—53.0 ± 8.0
2—84.0 ± 9.0
Nakagawa; 2007 (*) [19]HGF (I)
PGIS (II)
Plasmid (jet injector)Unspecified400 µgSham plasmid7 daysMcFarlane40 Sprague–Dawley male rats and 40 GK/Jcl male ratsIntracutaneous injection − 8 or 4 sites (1, 2) three days before flap elevation1—43.73 ± 12.78
2—44.42 ± 10.02
I + II1—44.76 ± 9.17
I + II2—60.24 ± 7.45
I—37.96 ± 10.15
II—32.2 ± 11.64
I + II—58.47 ± 12.15
Rah; 2014 (*) [20]HGFVirusConstant (up to 10 days)1 × 107 PFUPBS (A)
Sham protein (B)
10 daysMcFarlane30 Sprague–Dawley male rats (300–350 g)Subdermal injection two days and immediately after flap elevationA—39.2 ± 13.0
B—63.8 ± 6.4
71.4 ± 5.9
Huemer; 2004 [21]TGF-βVirusConstant (up to 7 days)1 × 108 PFUSaline (A)
Sham virus (B)
7 daysEpigastric30 Sprague–Dawley male rats (300–400 g)Subdermal injection just prior to flap elevationA—82.6 ± 4.6
B—82.2 ± 8.7
90.3 ± 4.0
Huemer; 2005 [22]TGF-β (I)
Shockwave (II)
VirusConstant (up to 7 days)1 × 108 PFU No treatment7 daysEpigastric30 Sprague–Dawley male rats (300–400 g)Subdermal injection just prior to flap elevation82.6 ± 4.3I—90.3 ± 4.0
II—97.7 ± 1.8
Liu; 2009 (*) [23]FGF-βVirusUnspecified7.5 × 1010 PFUSaline 7 daysMcFarlane38 Sprague–Dawley female rats (250–300 g)Intradermal injection 14 days, 7 days, or at the time of flap elevation (1, 2, 3)50.9 ± 5.21—56.1 ± 11.8
2—42.7 ± 8.4
3—57.6 ± 3.3
Lee; 2011 [24]RelaxinVirusConstant (up to 10 days)1 × 107 PFUPBS (A)
Sham virus (B)
10 daysMcFarlane30 Sprague–Dawley male rats (300–350 g)Subdermal injection two days before and immediately after flap elevationA—37.0 ± 7.0
B—43.0 ± 3.0
55.0 ± 5.0
Jung; 2003 [25]Ang1VirusUnspecified1 × 108 PFU No treatment (A)
Sham virus (B)
7 daysEpigastric19 Sprague–Dawley male rats (200–250 g)Subdermal injection two days before flap elevationA—67.76 ± 5.98
B—78.65 ± 6.44
84.24 ± 6.54
Choi; 2020 [26] DKK2VirusConstant (up to 10 days)1 × 107 PFUNo treatment (A)
Sham virus (B)
10 daysMcFarlane30 Sprague–Dawley male rats (300–350 g)Subdermal injection two days before and immediately after flap elevationA—no results
B—57.5 ± 4.21
80.0 ± 4.49
Lou; 2021 [27]PLA2G4EVirusConstant (up to 7 days)5 × 109 PFUSham virus (measured here)7 daysMcFarlane191 C57BL/6 J male mice (20–30g)Subcutaneous injection of viral particles in 3 areas, 28 days prior to flap elevation56.81 ± 8.0381.57 ± 4.87
Gurunluoglu; 2002 (*) [28]VEGF-164VirusUnspecified108 PFUSaline (A)
Sham virus (B)
7 daysEpigastric84 Sprague–Dawley male rats (300–350 g)Subdermal injections, 12 h, 3 days, 7 days, and 14 days prior to flap elevation (1, 2, 3, 4)A1—68.3 ± 4.3
B1—72.3 ± 13.1
A2—65.7 ± 3.2
B2—78.7 ± 6.9
A3—63.8 ± 7.0
B3—70.9 ± 13.9
A4—69.6 ± 5.3
B4—68.5 ± 10.1
1—86.0 ± 5.5
2—88.7 ± 2.4
3—89.3 ± 5.3
4—86.9 ± 4.6
Gurunluoglu; 2005 (*) [29]VEGF-121VirusConstant (up to 21 days) 108 PFUSaline (A)
Sham virus (B)
7 daysPeninsular abdominal flap34 Sprague–Dawley male rats (300–500 g)Subdermal injection at the time of flap elevationA—65.6 ± 9.4
B—56.0 ± 3.4
88.9 ± 6.1
Giunta; 2005 (*) [30]VEGF-165VirusConstant (up to 7 days)5 × 108 PFU 1 × 109 PFU(X)Saline7 daysAbdominal random-pattern50 Sprague–Dawley male rats (350 g)Subcutaneous injection at the time of flap elevation, 3 or 7 days before (1, 2, 3)1—24.65 ± 2.271—29.0 ± 6.39
2—45.19 ± 10.32
3—48.5 ± 4.12
3 X—60.81 ± 6.25
Fujihara; 2005 (*) [31]FGF-βPlasmidConstant (up to 3 days)300 µgSham plasmid7 daysDorsal island skin52 Sprague–Dawley male rats (200–250 g)Intramuscular injection with additional electroporation (E) two days before flap elevation71.9 ± 2.5
E—71.7 ± 4.1
70.3 ± 3.3
E—78.3 ± 5.3
Ferraro; 2009 (*) [32]VEGF-165PlasmidTransient (at 12 days baseline expression)100 µg No treatment (A)
Sham plasmid (B)
14 daysSingle pedicle random51 Sprague–Dawley male rats (275–300 g)Intradermal injection (group with additional electroporation, E) two days before flap elevationA—73.5 ± 4.1
BE—70.6 ± 7.4
65.6 ± 9.3
E—95.2 ± 2.2
De Freitas; 2010 (*) [33]VEGF-165Plasmid Unspecified100 µgNo treatment (A)
PBS (B)
Sham plasmid (C)
5 daysMuscle32 Wistar male rats (350–400 g)Intrafascial injection 30 days before flap elevation (during abdominoplasty) with electroporation in plasmid presenceA—75.35 ± 18.13
B—37.51 ± 28.06
C—42.2 ± 25.43
81.67 ± 16.20
Chen; 2011 (*) [34]VEGF-165Cells (transfection)Transient (at 14 days baseline expression)5 × 106 cellsNon-transfected cells7 daysRectangular full-thickness64 Wistar male and female rats (250–300 g)Subdermal injection with internal control at different times before flap elevation (0, 2, 4, and 7 days—1, 2, 3, 4)1—36.57 ± 5.28
2—35.43 ± 4.73
3—34.54 ± 4.68
4—35.62 ± 4.51
1—62.63 ± 8.53
2—67.00 ± 7.19
3—71.44 ± 5.56
4—53.71 ± 5.34
Chang; 2021 [35]HIF1αPlasmidTransient (decrease after 48 h) 50 µgSham plasmid7 days (1) 14 days (2)McFarlane20 Sprague–Dawley male ratsIntradermal injection seven days before flap elevation1—68.75 ± 36.07
2—80.31 ± 21.25
1—92.97 ± 40.02
2—92.58 ± 15.07
Basu; 2014 [36]VEGF-165PlasmidUnspecified50 µgNo treatment7 days (1) 14 days (2)McFarlane109 Sprague–Dawley male rats (275–300 g)Intradermal injection at the time of flap elevation. Additional electrode array (E) during procedure.1—84.75 ± 6.8
1E—84.75 ± 6.8
2E—79.8 ± 5.2
1—88.7 ± 4.84
1E—98.48 ± 1.87
2E—98.22 ± 2.82
Antonini; 2007 (*) [37]VEGF-165VirusUnspecified150 µLSham virus14 daysEpigastric (Epi) Muscle (M)48 Wistar male rats (260–290 g)Subcutaneous/intramuscular injection at the time of flap elevation, 7 or 14 days prior (1, 2, 3)Epi1—63.9 ± 0.6
Epi2—61.1 ± 2.2
Epi3—62.8 ± 1.4
M1—77.3 ± 1.2
M2—74 ± 3.0
M3—75.4 ± 0.8
Epi1—72.2 ± 1.6
Epi2—76.7 ± 1.5
Epi3—78.3 ± 2.8
M1—77.6 ± 1.1
M2—83.9 ± 1.7
M3—87.7 ± 0.8
Taub;1998 (*) [38]VEGF-121PlasmidUnspecified7 µgSaline (A)
Sham plasmid (B)
7 daysEpigastric30 Sprague–Dawley female rats (250–300 g)Intra-arterial injection during flap elevationA—31.9 ± 12.64
B—28.1 ± 10.13
93.9 ± 4.91
Rinsch; 2001 (*) [39]VEGF-121 (I) VEGF-165 (II)
FGF-β (III)
Cells (transfection)Constant5 × 105 cellsNo treatment (A)
Capsule only (B)
Capsule containing non-modified cells (C)
7 daysMcFarlane 86 Wistar female rats (250–300 g)Administration of capsule on the subcutaneous tissue of flap during elevationA—50.0 ± 3.8
B—44.9 ± 7.8
C—53.2 ± 5.8
I—47.2 ± 11.3
II—46.9 ± 9.7
III—65.8 ± 6.3
O’Toole; 2002 (*) [40]VEGF-165 (I) VEGF-167 (II) VEGF-186 (III)PlasmidConstant (up to 2 days)50 µgSham plasmid (A)
Saline (B)
7 daysEpigastric60 Sprague–Dawley male rats (400–550 g)Subcutaneous injection at the time of flap elevationA—83.2 ± 12.08
B—81.2 ± 12.08
I—90.3 ± 8.4
II—92.1 ± 7.09
III—86.2 ± 6.57
Yang; 2005 (*) [41]VEGF-121PlasmidUnspecified80 µgSham plasmid (A)
Saline (B)
7 daysMcFarlane30 Sprague–Dawley female rats (280–320 g)Intramuscular injection at the time of flap elevationA—47.0 ± 6.0
B—46.0 ± 5.0
87.0 ± 5.0
Zacchigna; 2005 (*) [42]VEGF-165VirusUnspecified1.5 × 1011 PFUSham plasmid7 daysEpigastric (Epi) Muscle (M)88 Wistar male rats (250–300 g)Subcutaneous (E) or intramuscular (M) injection at the time of flap elevation (1), 7 days before (2) or 14 days before (3)1E—62.6 ± 1.8
2E—61.0
3E—63.0
1M—78.0
2M—75.0
3M—77.2 ± 2.7
1E—71.0
2E—77.0
3E—79.0
1M—78.0
2M—85.0
3M—91.1 ± 2.8
Zhang; 2005 (*) [43]VEGF-165Plasmid Unspecified50 µgSham plasmid (A)
Saline (B)
4 daysMuscle 52 Sprague–Dawley rats (380–420 g)Subcutaneous injection 4 days before flap elevationA—36.3 ±13.1
B—43.4 ± 11.3
63.7 ± 20.2
Wang; 2006 (*) [44]PDGF-BPlasmidUnspecified50 µgSaline7 daysMcFarlane20 Sprague–Dawley female rats (250–300 g)Intradermal injection at the time of flap elevation52.4 ± 5.160.1 ± 7.2
Yi; 2006 (*) [45]VEGF-165Cells (transfection)Transient (peak at day 4)5 × 105 cellsNon-transfected cells (A) culture medium (B) 28 daysCranially based30 athymic nude miceSubcutaneous injection three days before flap elevationA—59.37 ± 14.83
B—40.0 ± 16.65
68.16 ± 20.4
Zheng; 2008 (*) [46]VEGF-165Cells (transfection)Transient (baseline expression at day 14)5 × 106 cellsNon-transfected cells (A) culture medium (B)14 daysMcFarlane30 Sprague–Dawley rats (100–120 g)Subcutaneous injection four days before flap elevationA—66.4 ± 6.1
B—51.5 ± 7.5
83.1 ± 2.6
Spanholtz; 2009 (*) [47]VEGF-165Cells (transduction)Transient (baseline expression at day 5)1 × 107 cellsSham transduced cells (A)
Non-transduced cells (B) saline (C)
7 days McFarlane80 Sprague–Dawley female rats (200–225 g)Intradermal injection at different times before flap elevation: at the same time (1), 7 days (2), or 14 days before (3). Injection within the flap and into the surroundingsA—44.05 ± 3.92
B—45.0
C—47.0
1—47.60 ± 5.11
2—54.01 ± 5.95
3—46.54 ± 5.02
Rezende; 2010 (*) [48]VEGF-165PlasmidUnspecified50 µgEmpty plasmid in two different areas (A, B), sham plasmid (C)
Saline (D)
5 daysMuscle49 Wistar male rats (300 g)Intradermal injection at the time of flap elevation with electroporation in different areas (* or **)A—77.20 ± 9.79
B—61.38 ± 12.50
C—no data
D—75.0 ± 1.0
*—60.77 ± 27.10
**—33.06 ± 31.76
Spanholtz; 2011 (*) [49]FGF-β (I) VEGF-165 (II)Cells (transfection)Transient (baseline expression at day 14)5 x106 cells (*) 1 × 107 cells(**)Sham-modified fibroblasts (compared here)
Non-modified fibroblast
Cell medium only
14 daysMcFarlane320 Sprague–Dawley female rats (200–225 g)Subdermal injections 7 days or 14 days (1, 2) before flap elevation within flap alone (*) and in flap surrounding and flap (**)1 *—51.02 ± 2.96
2 *—51.90 ± 1.43 1 **—52.79 ± 3.01 2 **—51.56 ± 1.43
I + II * 1—58.76 ± 1.70
I + II * 2—60.38 ± 2.77
I + II ** 1—59.20 ± 2.17
I + II ** 2—62.16 ± 1.38
I * 1—53.48 ± 1.72
I * 2—54.22 ± 2.21
I ** 1—60.53 ± 1.68
I ** 2—57.57 ± 2.15
Wang; 2011 (*) [50]VEGF-165 VirusUnspecified3 × 1010 PFUSham virus (A)
Saline (B)
7 days McFarlane30 Sprague–Dawley female rats (250–300 g)Intradermal injection 14 days before flap elevationA—45.9 ± 6.3
B—48.7 ± 4.9
55.8 ± 6.9
Uemura; 2012 [51]NF-kbOligonucleotideTransient1–2 mgSham oligonucleotide (A)
No treatment (B)
5 daysEpigastric36 Sprague–Dawley male rats (350–400 g)Intra-arterial injection at the time of flap elevationA—31.1 ± 3.7
B—31.7 ± 4.8
57.9 ± 8.4
Wang; 2013 [52]KGFVirusUnspecified1 × 109 PFUSham virus with dexamethasone (A)
Dexamethasone (B)
PBS (C)
35 daysMcFarlane60 Sprague–Dawley male rats (300–350 g)Subdermal injection in wound margin five days after flap elevationA—86.19 ± 1.63
B—85.33 ± 1.98
C—84.20 ± 2.55
88.63 ± 0.69
Jafari; 2017 (*) [53]HGFPlasmidUnspecified25 µgNo treatment7 daysMcFarlane15 Wistar male rats (290–320 g)Intradermal injection 24 h before (1) or 24 h after (2) flap elevation followed by electroporation64.67 ± 3.901—72.86 ± 7.46 2—2.93 ± 4.81
Jafari; 2018 [54]IL-10PlasmidUnspecified100 µgNo treatment7 daysMcFarlane9 Wistar male rats (300–330 g)Intradermal injection 24 h before flap elevation followed by electroporation64.77 ± 3.9081.26 ± 4.70
Jafari; 2021 [55]IL-10 (I)
HGF (II)
VEGF-165 (III)
PlasmidUnspecified100 µgNo treatment 7 days McFarlane15 Wistar male rats (290–320 g)Intradermal injection 24 h before flap (1) or 24 h (2) after flap elevation followed by electroporation64.77 ± 3.90I1 + III2—81.66 ± 9.70
I1 + II2—74.06 ± 1.65
Zhang; 2011 [56]SDF-1Cells (transduction) (I)
Virus (II)
Unspecified1 × 106 cells (I)
6.25 × 109 PFU (II)
Non-transduced MSC (A)
saline (B)
10 daysEpigastric24 Lewis rats (200–300 g)Intravascular injection proximal to the anastomosis of
femoral artery at the time of flap elevation
A—88.56 ± 3.72
B—79.12 ± 3.34
I—96.82 ± 2.35
II—86.86 ± 4.09
Luo; 2021 [57]SDF-1αCells (modRNA transfection)Constant (up to 14 days)5.4 × 107 cellsSham transfected fibroblasts (A)
PBS (B)
10 days (extended to 28 days for the treatment group)McFarlane60 Sprague–Dawley male rats (250–300 g)Intradermal injection at the time of flap elevationA—animals died
B—animals died
after 28 days—92.2
Legend: VEGF—vascular endothelial growth factor, Shockwave—extracorporeal shockwave therapy, PDGF—platelet-derived growth factor, FGF-β—fibroblast growth factor 2, eNOS—endothelial nitric oxide synthase, HGF—hepatocyte growth factor, PGIS—prostacyclin synthase, TGF-β—transforming growth factor beta, Ang1—angiopoietin 1, DKK2—Dickkopf-related protein 2, PLA2G4E—phospholipase A2, HIF1α—hypoxia-inducible factor 1-alpha, NF-kb—nuclear factor kappa-light-chain-enhancer of activated B cells, KGF—keratinocyte growth factor, IL-10—interleukin 10, PFU—plaque-forming unit, PBS—phosphate-buffered saline, SDF-1—stromal cell-derived factor 1, MSC—mesenchymal stem cells, modRNA—modified RNA.
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

Paskal, W.; Gotowiec, M.; Stachura, A.; Kopka, M.; Włodarski, P. VEGF and Other Gene Therapies Improve Flap Survival—A Systematic Review and Meta-Analysis of Preclinical Studies. Int. J. Mol. Sci. 2024, 25, 2622. https://doi.org/10.3390/ijms25052622

AMA Style

Paskal W, Gotowiec M, Stachura A, Kopka M, Włodarski P. VEGF and Other Gene Therapies Improve Flap Survival—A Systematic Review and Meta-Analysis of Preclinical Studies. International Journal of Molecular Sciences. 2024; 25(5):2622. https://doi.org/10.3390/ijms25052622

Chicago/Turabian Style

Paskal, Wiktor, Mateusz Gotowiec, Albert Stachura, Michał Kopka, and Paweł Włodarski. 2024. "VEGF and Other Gene Therapies Improve Flap Survival—A Systematic Review and Meta-Analysis of Preclinical Studies" International Journal of Molecular Sciences 25, no. 5: 2622. https://doi.org/10.3390/ijms25052622

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop