Abstract
Background
   As an original traditional Chinese medicinal formula, Qin Huang formula
   (QHF) is used as adjuvant therapy for treating lymphoma in our hospital
   and has proven efficacy when combined with chemotherapy. However, the
   underlying mechanisms of QHF have not been elucidated.
Methods
   A network pharmacological-based analysis method was used to screen the
   active components and predict the potential mechanisms of QHF in
   treating B cell lymphoma. Then, a murine model was built to verify the
   antitumor effect of QHF combined with Adriamycin (ADM) in vivo.
   Finally, IHC, ELISA, ^18F-FDG PET-CT scan, and western blot were
   processed to reveal the intriguing mechanism of QHF in treating B cell
   lymphoma.
Results
   The systemic pharmacological study revealed that QHF took effect
   following a multiple-target and multiple-pathway pattern in the human
   body. In vivo study showed that combination therapy with QHF and ADM
   potently inhibited the growth of B cell lymphoma in a syngeneic murine
   model, and significantly increased the proportion of tumor infiltrating
   CD4+ and CD8+ T cells in the tumor microenvironment (TME). Furthermore,
   the level of CXCL10 and IL-6 was significantly increased in the
   combination group. Finally, the western blot exhibited that the level
   of TLR2 and p38 MAPK increased in the combination therapy group.
Conclusion
   QHF in combination of ADM enhances the antitumor effect of ADM via
   modulating tumor immune microenvironment and can be a combination
   therapeutic strategy for B cell lymphoma patients.
Supplementary Information
   The online version contains supplementary material available at
   10.1186/s12906-022-03660-8.
   Keywords: Qin Huang formula, B-cell lymphoma, Tumor microenvironment,
   Tumor-infiltrating lymphocytes, Toll-like receptor, Systemic
   pharmacology
Introduction
   Non-Hodgkin lymphoma (NHL) is a malignancy that mainly infringes the
   lymphoid system, with extranodal sites often affected as well. It has
   arisen to the top 10 most common cancers in 2020 [[41]1]. Among all the
   subtypes of NHLs, diffuse large B cell lymphoma (DLBCL) is the most
   common subtype and accounts for 25-30% of diagnoses [[42]2]. Albeit
   aggressive, most DLBCL is chemo-sensitive, and R-CHOP (rituximab,
   cyclophosphamide, doxorubicin, vincristine, and prednisone) is the
   standard regimen. However, due to resistance to R-CHOP, there are still
   ~ 30% of patients suffering from refractory or relapse (r/r) disease,
   and their prognosis is rather poor [[43]3, [44]4]. Such treatment
   plight has prompted the emergence of novel treatment options, such as
   therapeutics targeting immune checkpoints, tumor microenvironment
   (TME), signaling pathways, and cellular immunotherapy [[45]5]. As a
   cellular immunotherapy, chimeric antigen receptor (CAR) T-cell therapy
   has shown a promising future in the management of B cell lymphomas. Two
   CAR T-cell products are now approved for the treatment of
   relapsed/refractory large B-cell lymphoma [[46]6]. However, its acute
   and late toxicities, as well as on-target off-tumor effect should be
   addressed [[47]7]. PD-1/PD-L1 blockade, as an immune checkpoint
   inhibitor (ICI), aims to protect cytotoxic T cells and restore their
   function from PD-1/PD-L1 mediated immune evasion. However, its efficacy
   in DLBCL remains unsatisfactory compared to classical Hodgkin Lymphoma
   (cHL) [[48]8, [49]9]. Research accumulated that the pre-existing immune
   landscape within the TME may influence the response to immunotherapies
   [[50]10–[51]12] and R-CHOP [[52]13, [53]14]. Therefore, it is pivotal
   to explore agents that can modulate TME. TME is an extremely intricate
   environment that harbors diverse cells, structures, and matters, with
   one component interacting with many others, forming complex regulatory
   networks. Drugs that have multiple targets might be a promising
   combination therapy to modulate TME, especially those that could elicit
   both innate and adaptive immune responses against cancers.
   Traditional Chinese medicine (TCM) has a long history in treating
   various diseases and ailments. Formula, a prescription that contains
   multiple drugs with different but related modes of action, is a
   representative of combination therapy in the system of TCM [[54]15].
   With the guidance of this philosophy, the Qin Huang formula (QHF), was
   grouped and used as adjuvant therapy for treating lymphoma in our
   hospital (Patent No.:2015103646392). Containing Scutellaria baicalensis
   Georgi. (Lamiaceae) (SR), Astragalus membranaceus (Fisch) Bunge (AM),
   Prunella vulgaris L. (Lamiaceae) (PV), and Curcuma Longa L.
   (Zingiberaceae) (CL), the 4-herb formula conforms to the theology of
   TCM in treating malignancies. Huang et al. [[55]16] found that
   baicalin, an ingredient of SR, could down-regulate PI3K/Akt signaling
   pathway and induce apoptosis in the Burkitt lymphoma cell line. Kumagai
   et al. [[56]17] demonstrated that SR and its major components,
   baicalein, and wogonin, could exert anti-proliferation and induce
   apoptosis mediated by mitochondrial damage in lymphocytic leukemia,
   Burkitt lymphoma, and myeloma cell lines. Studies [[57]18–[58]20] also
   showed that PV and CL have a strong antiproliferation effect upon Raji,
   a Burkitt lymphoma cell line, and other solid tumors. Meanwhile, it is
   reported that AM and its extract could protect human epithelial and
   endothelial cells from LPS-induced apoptosis [[59]21] and exert a
   cardioprotective effect against Adriamycin-induced cardiotoxicity
   [[60]22]. Thus, as a mixture of the four herbs, QHF was expected to
   exert an antitumor effect in B cell lymphoma as well. And our previous
   work [[61]23] found that QHF (also referred to as Qin Huang mixture)
   has a modulating effect on the systemic immune function of lymphoma
   patients who have completed R-CHOP chemotherapy. Besides, we also found
   that when combined with R-CHOP, QHF can significantly lower the overall
   side effect frequencies and improve the short-term effects of R-CHOP in
   treating initial onset DLBCL patients [[62]24]. In addition, we
   explored the effects of several major flavonoids of QHF in vitro and
   found that wogonin, luteolin, kaempferol, quercetin, and silymarin can
   inhibit the proliferation and induce apoptosis of the Raji, SU-DHL-4,
   and A20 cell lines (data not published), which is consistent with other
   studies [[63]25–[64]28]. Therefore, we hypothesized that QHF can
   enhance the antitumor effects of chemotherapy by modulating immune
   functions, especially the tumor immune microenvironment (TIME), of B
   cell NHL. In this study, a systemic pharmacological analysis was
   carried out to predict the targets and mechanisms of QHF in treating B
   cell lymphoma. Then, the antitumor effect of QHF in combination with
   Adriamycin (ADM) was evaluated on a murine model induced by the mouse B
   cell lymphoma cell line A20. Moreover, the TME of the mouse B cell
   lymphoma was further evaluated via IHC staining, ELISA, as well as
   western blot to confirm our hypothesis. Our extensive study provides
   experimental evidence regarding the antitumor effect of QHF + ADM
   combination therapy and revealed its underlying mechanisms.
Methods
Screening the chemical constituents of QHF and their targets
   The ingredients information of QHF was searched through four databases:
   TCMSP [[65]29] ([66]https://www.tcmspw.com/tcmsp.php), DrugBank
   [[67]30] ([68]https://go.drugbank.com/#), BATMAN-TCM [[69]31]
   ([70]http://bionet.ncpsb.org.cn/batman-tcm/), and ETCM [[71]32]^30
   ([72]http://www.tcmip.cn/ETCM/). These databases contain information on
   chemical constituents and their targets of herbs. The screening
   criteria were set as oral bioavailability (OB) ≥30% and drug-likeness
   (DL) > 0.18 to collect the potential ingredients and their targets of
   SR, AM, PV, and CL.
   B cell lymphoma-associated targets were collected from the following
   databases: Genecards [[73]33] ([74]https://www.genecards.org/), GEO
   [[75]34] ([76]https://www.ncbi.nlm.nih.gov/geo/), DisGeNET [[77]35]
   ([78]https://www.disgenet.org/), and OMIM [[79]36]
   ([80]https://www.omim.org/). As of GEO analysis, the [81]GSE12453
   dataset was utilized to draw the differentially expressed genes of
   DLBCL compared with naive B cells. The results with P-adjusted value
   < 0.05 and | log2(fold change) | > 1 were kept for further analysis.
   Then R platform ggplot2 [[82]37] package was used to draw a volcano
   plot. Finally, with the help of the R platform VennDiagram [[83]38]
   package, the common targets of QHF and B cell lymphoma were analyzed.
The protein-protein interaction (PPI) network and drug-disease-target network
building
   The common targets were imported to the STRING [[84]39] database
   (version 11.0, [85]https://string-db.org) to construct the PPI network.
   The gene expression was correlated with others by minimum required
   interaction score thresholds of 0.9. Disconnected nodes in the network
   were hidden. The nodes in the PPI network represent the protein
   targets, while the edges stand for the interaction between two targets.
   Then, topological parameters of nodes betweenness, closeness, degree,
   network centrality, as well as local clustering coefficient were
   analyzed via R in the network. Targets with node degrees over twice the
   average were kept and ranked by node degree. Then the top 30 targets
   were counted to be the core targets. Circlize [[86]40] package (version
   0.4.13) of the R platform [[87]41] was used to construct an herb-target
   chord plot to display the relationship between the herbs and the core
   targets. And a drug-disease-target network was constructed with the
   utilization of Cytoscape 3.6.0 software [[88]42] to show the
   interactions of the compounds and the core targets.
Gene ontology (GO) functional and Kyoto encyclopedia of genes and genomes
(KEGG) pathway enrichment
   GO and KEGG enrichment analysis was employed to further clarify the
   biological interpretations and signaling pathways of the 30 core
   targets we obtained in Part 2.2. ClusterProfiler [[89]43], an R
   (version 4.1.0) package for GO and KEGG enrichment analysis tool, was
   utilized to access the databases of GO and KEGG. Pathview [[90]44]
   package of R platform was applied to visualize KEGG pathways. The genes
   with adjusted P-value < 0.05 were retained.
The molecular docking processing
   Although the relationship between the targets and compounds was
   verified, the intensity of the interaction remained unclear. The
   computer-assistant molecular docking technology was used in this study
   to further investigate the ligation intensity. First, the SDF files of
   the compound structures were downloaded from the PubChem database
   ([91]https://pubchem.ncbi.nlm.nih.gov/). Second, the protein data bank
   (PDB) ([92]http://www.rcsb.org/) database was searched to obtain the
   molecular structures of the target proteins. The ligand-receptor
   compound structures were priorly chosen and modified by removing the
   original ligands. The pocket of the combination was constructed in the
   slot. Third, after adding polar hydrogen atoms, the AutoDock Vina
   software [[93]45] (ver. 1.1.2, the Scripps Research Institute, U.S.)
   was used to achieve the binding free energy of ligands and receptors.
   Finally, the bonds within the ligand-receptor complexes were visualized
   by Ligplot software [[94]46] (ver. 4.5.3, EBI, UK).
Materials and reagents
   The herbal formula QHF is constituted of four medicinal herbs: SR
   (20 g), AM (20 g), PV (15 g), and CL (10 g) at a rate of 4:4:3:2
   (w/w/w/w). The herbs were obtained from Shanghai Wanshicheng
   Pharmaceutical Co. Ltd. (Lot Number: SR 20210406-1; AM 20210515-1; PV
   20210604-1; CL 20210526-1), and the frozen powder of QHF was prepared
   by Shanghai Traditional Chinese Medicine Technology Co. Ltd. To obtain
   the extract of QHF, the herbs were first soaked in water at tenfold
   volume (v/w) overnight, then extract the decoction twice for 1 h each
   time with a tenfold volume of water to herbs (v/w). After filtration,
   the solution was evaporated under reduced pressure at 60 °C to a
   relative density of 1.15, then the extract was frozen desiccated to
   powder, and stored at − 20 °C for further use.
   According to the pre-study of the effective dosage of QHF we conducted
   on mice, we found QHF (6000 mg/kg) is more effective compared with QHF
   (300 mg/kg) and QHF (3300 mg/kg), but tolerable as well (see Additional
   file [95]1). Therefore, the regimen of the combination group (QHF
   (6000 mg/kg) + ADM (5 mg/kg)) was used in this study.
Animals and treatments
   Nineteen male Balb/c mice aged 6-8 weeks were purchased from the
   Animals Care Centre at Shanghai Jiao Tong University. The animals were
   given free access to food and water and acclimatized for 1 week under
   standard conditions and 12 h light/dark cycle. All experimental
   procedures were conducted under the requirements of the research ethics
   Committee at Shanghai Jiao Tong University (Ethical reference no.
   10687).
   The mouse B cell lymphoma A20 cell line was kindly provided by the
   Institute of Hematology, Shanghai Ruijin Hospital, and cultured in RPMI
   1640 medium (Gibco, Gaithersburg, MD, USA) containing 10% fetal bovine
   serum (FBS), 100 U/mL penicillin, and 100 μg/ mL streptomycin. The
   cells were incubated at 37 °C with 5% CO[2]. Then the cells
   (~ 1.0 × 10^7 cells/mouse) were transplanted subcutaneously into the
   right axillary region of the mice. After 1 week or so, when the tumors
   reached a size of 50 mm^3, the mice were randomly allocated into three
   groups as follows:
     * Control group (Control): intravenously injected with 0.1 mL PBS
       three times on day 1, day 4, and day 8, respectively.
     * Adriamycin group (ADM): received ADM (5 mg/kg) intravenously (i.v.)
       three times on day 1, day 4, and day 8, respectively [[96]47].
     * Combination therapy group (QHF + ADM): received 6000 mg/kg QHF
       dissolved in ddH[2]O intragastrically, 0.1 mL per day; ADM was
       administered same as ADM group.
   The tumor diameters were measured routinely with a caliper. Tumor
   volume was estimated as follows: length × width^2× 0.5. Besides, the
   tumor growth inhibition rate (TGI, %) was calculated to evaluate the
   anticancer effect of the two regimens.
   [MATH: TGI%=1-tumorvolum
   eoftreat
   edgroup
   tumorvolum
   eofcontr
   olgroup
   *100% :MATH]
   QHF was administered intragastrically (i.g.) every day throughout the
   experiment. On day 12, the mice were sacrificed under euthanasia with
   pentobarbital sodium. After dissection, the tumor of each mouse was
   divided into two parts, one part was kept in 4% paraformaldehyde and
   the other was freshly frozen in liquid nitrogen and then stored at
   − 80 °C.
Immunohistochemistry (IHC) on formalin-fixed paraffin-embedded samples
   Tumors were embedded in paraffin blocks and cut into tissue sections
   4 μm thick. Then hematoxylin & eosin staining was processed to confirm
   the presence of the tumors. Besides, paraffin sections were dewaxed
   with xylene and rehydrated with alcohol at graded concentrations for
   immunohistochemical staining. Endogenous peroxidase was inactivated
   with 3% H[2]O[2] for 15 minutes. Then, the slides were blocked with 5%
   goat serum for 20 min at 37 °C, followed by incubated with anti-CD4
   (1:3000, Servicebio, China), anti-CD8 (1:1000, Servicebio, China),
   anti-CD206 (1:500, Servicebio, China) and anti-iNOS (SP126, 1:100,
   Abcam, UK) antibodies overnight at 4 °C. The next day, samples were
   incubated with a secondary antibody (Servicebio, China) for 1 hour at
   room temperature. After that, staining the samples with the
   ready-to-use reagent DAB kit. Finally, the sections were observed with
   a microscope.
   The images of the tissue sections were obtained by the tissue slice
   digital scanner. The Servicebio image analysis system was used to
   automatically analyze and calculate the number of weak, medium, and
   strong positive cells in the measurement area (negative for no
   coloring, score 0 points; weak positive for light yellow, score 1
   point; medium positive for brownish yellow, score 2 points; strong
   positive for tan, score 3 points). The positive cell rate indicator was
   used to assess the percentage of positive cells [[97]48].
   [MATH: positivecellrate%=numberofposit
   ivecells
   numbe
   roftotal
   cells
   *100%. :MATH]
Enzyme-linked immunosorbent assay (ELISA) on FFPE samples
   The total protein was extracted from the freshly frozen tumor tissues
   previously stored. First, 1 ml of RIPA buffer was prepared for every
   0.1 g of tissue and protease and phosphatase inhibitors were added to a
   final 1x concentration. The tissues were ground under 60 Hz for
   4 cycles. Then, the homogenate was centrifuged at 12500 rpm at 4 °C for
   15 minutes. After this, the supernatant was transferred to a clean tube
   for ELISA. According to the manufacturer’s instructions, commercially
   available ELISA kits (MultiSciences (LIANKE) Biotech Co., Ltd.,
   Hangzhou, China) were applied for mouse protein expression detection of
   CXCL10, IL-2, IL-17, IL-6, TGF-β, and IFN-γ levels.
Western blotting
   Three freshly frozen samples of each group were also homogenized in
   RIPA buffer supplemented with proteinase/phosphatase inhibitors. After
   centrifuging, the protein concentration was determined with a BCA
   protein assay kit (Beyotime, Shanghai, China). Twenty μg protein from
   each sample was subjected to 12% SDS-PAGE and electrotransferred to
   0.22uM PVDF membranes. Then, the membranes were subjected to blocking
   buffer (Beyotime, Shanghai, China), followed by incubation overnight at
   4 °C with primary antibodies (1:1000, Abcam, U.S.) against p38 MAPK, p-
   ERK, TLR2, and β-actin. After that, the probed membranes were washed
   with TBST three times, followed by incubation with secondary antibodies
   (1:5000, Abcam, U.S) for 1 h at room temperature. The protein signals
   were detected using a chemiluminescence ECL assay kit and imaged using
   a Tanon 5200 imaging system. β-actin was served as the internal
   reference. The expression levels of proteins were calculated by Image J
   platform.
^18F-FDG PET-CT scan
   One mouse of each group was picked out to receive the ^18F-FDG PET-CT
   scanning on the first day (d1) and the last day (d8) of therapy,
   respectively. The combination group underwent another ^18F-FDG PET-CT
   scan on the 7th day after discontinuing therapy. PET/CT imaging was
   performed on the Inveon MM Platform (Siemens Preclinical Solutions,
   Knoxville, Tennessee, USA), which was equipped with a
   computer-controlled bed and 8.5 cm transaxial and 5.7 cm axial fields
   of view (FOV). After fasting overnight for 8 h and being anesthetized
   under pentobarbital sodium, three mice received an intravenous infusion
   of 100-200 uCi ^18F-FDG. The scanning process was performed with Inveon
   Acquisition Workplace (IAW) 1.5.0.28. Before the PET scan, the CT X-ray
   for attenuation correction was scanned for 10 minutes with a power of
   80 Kv and 500 uA and an exposure time of 1100 ms. Then, 10-minute
   static PET scans were acquired, followed by images reconstruction with
   an OSEM3D (Three-Dimensional Ordered Subsets Expectation Maximum)
   algorithm. The 3D regions of interest (ROIs) were drawn over the right
   axillary region guided by CT, with the tracer uptake measured by the
   Inveon Research Workplace (IRW) 3.0. Individual quantification of the
   ^18F-FDG uptake, as well as the max uptake values (SUV[max]) of the
   mice, were determined.
Statistical analysis
   The obtained data were expressed as mean ± standard errors. Statistical
   analysis was performed on the platform of GraphPad Prism 9 (GraphPad
   Software, San Diego, CA, USA). Comparisons among multiple groups were
   performed by one-way ANOVA with Tukey’s multiple comparisons test.
   Comparisons between two groups were assessed using Student’s unpaired
   t-tests. A p-value < 0.05 was considered significant.
Results
Active components and hub genes of QHF in treating B cell lymphoma
   A total of 271 active ingredients of QHF with OB ≥ 30% and DL > 0.18
   were obtained. In the GEO database, the samples of [98]GSM312858-312869
   (DLBCL), [99]GSM312870-312876 (naive B-cells), and
   [100]GSM312877-312886 (memory B cells) from [101]GSE12453 were
   selected. The volcano plot shows the relationship between the
   significance and the foldchange of differentially expressed genes
   (DEGs). They were highlighted by the red and green dots (Fig. [102]1A).
   Filtered by p-adjusted value < 0.05, a total of 7141 transcriptionally
   dysregulated genes of B-cell lymphoma were discovered. The Venn diagram
   shows the overlapping of QHF (red color) and DLBCL (turquoise color)
   related targets (Fig. [103]1B). Finally, we obtained a total of 541
   common targets. To explore the interactions between the targets of QHF,
   a PPI network was constructed with the STRING database. After importing
   541 common targets into the database, a total of 539 nodes and 2807
   edges were built with an average node degree of 10.4 (Fig. [104]1C). By
   topology analysis of the interactions in the PPI network, we obtain a
   bar plot displaying the core targets and their node degrees (Fig.
   [105]1D). According to this figure, MAPK1/8/14, TP53, EP300, AKT1, APP,
   STAT3, JUN, RELA, TNF, IL6, C3, HSP90AA1, AGT, NFKB1, RAC1, NR3C1,
   EGFR, RXRA, ESR1, FOS, RHOA, APOB, RB1, SUMO1, FGA, NCOA1, PRKCD, and
   ANXA1 were the top 30 targets with the most nodes degree. Among these
   core targets, MAPK1/ERK2, MAPK8/JNK1, and MAPK14/p38 consist of the
   MAPK signaling pathway, which regulates diverse biological functions,
   including cell growth, differentiation, and survival. JUN is one of the
   components of AP-1, which can be phosphorylated by MAPK8/JNK1 and
   induce the expression of inflammatory cytokines, including IL-6 and
   TNF-α. NFKB and AKT1 are the key molecules in the NF-κB signaling
   pathway and PI3K-Akt signaling pathway, respectively. The above three
   pathways are the downstream pathway of TLRs.
Fig. 1.
   [106]Fig. 1
   [107]Open in a new tab
   Active ingredients and core targets screening. A. The volcano plot
   displayed the significance and the fold change of differentially
   expressed genes acquired from the GEO database; The red ones
   represented up-regulated DEGs (log2(fold change) > 1), while the green
   ones represented down-regulated DEGs (log2(fold change) < − 1). B. The
   Venn diagram demonstrates the overlapping of network pharmacology-based
   targets of QHF components (red color) and transcriptionally
   dysregulated genes of B-cell lymphoma (turquoise color). C. The
   Protein-Protein-Interaction (PPI) network of the core genes. D. The
   core targets and their node degrees. E. The herb-target chord chart
   displays the relationship of the most involved targets and herbs. F.
   The drug-disease-target network indicated the relationship between the
   drugs, compounds, and targets. The pink triangles of the left
   peripheral circle represented the compounds of QHF, while the four
   orange octagons represented the four herbs of QHF. The blue arrows of
   the right peripheral circle represented the core targets of B cell
   lymphoma
   To further investigate the relationship between herbs, components, and
   hub targets, the R platform was utilized to form a chord chart (Fig.
   [108]1E). According to the plot, there were 64, 96, 72, and 13 edges in
   AM, SR, PV, and CL, respectively. The herb which related to most
   targets was SR (degree = 96). In addition, the relationship between
   drugs and the disease was described by the drug-disease-target network
   (Fig. [109]1F). In this network, there were 220 nodes and 625 edges.
   Quercetin in AM and PV (degree = 20), kaempferol in AM and PV
   (degree = 12), luteolin in PV (degree = 9), and wogonin in SR
   (degree = 9) ranked the top four compounds that connected most quantity
   of targets of DLBCL. In CL, stigmasterol relates to the most targets in
   the network. Among these five active components, four belong to
   flavonoids and only stigmasterol of CL belongs to sterol. The
   structural formula and other important parameters of the major active
   components of QHF were shown in Table [110]1.
Table 1.
   The structural formula and other important parameters of the major
   active components of QHF.
   [111]graphic file with name 12906_2022_3660_Tab1_HTML.jpg
   [112]Open in a new tab
   ^*Note: SR for Scutellaria baicalensis Georgi. (Lamiaceae); AM for
   Astragalus membranaceus (Fisch) Bunge; PV for Prunella vulgaris L.
   (Lamiaceae); CL for Curcuma Longa L. (Zingiberaceae).
Predicted mechanisms of QHF in treating B cell lymphoma
   As mentioned above, the top 30 core targets (see Additional file
   [113]2) were input for GO enrichment and KEGG pathway enrichment
   analyses to show the biological activity and potential mechanisms of
   the hub genes. The most enriched functions in the GO analysis are shown
   in Fig. [114]2A. In detail, the top three terms in the GO biological
   processes were DNA-binding transcription factor binding (GO:0140297),
   RNA polymerase II-specific DNA-binding transcription factor binding
   (GO:0061629), DNA-binding transcription activator activity, RNA
   polymerase II-specific (GO:0033613).
Fig. 2.
   [115]Fig. 2
   [116]Open in a new tab
   Predicted mechanisms of QHF in treating B cell lymphoma. A. The dot
   plot of GO functional enrichment of QHF. B. The dot plot of KEGG
   pathway enrichment analysis of QHF. C. The KEGG pathway diagram of
   Toll-like receptor signaling pathway [[117]49–[118]51]. It shows that
   QHF could target AKT, NF-κB, p38, ERK, and JNK to activate PI3K/Akt
   signaling pathway, MAPK, and NF-κB signaling pathways. D.
   Representative images of molecular docking show that the active
   components of QHF bind to TLR2, ERK, JNK, and AKT primarily with
   hydrogen bonds
   KEGG pathways enrichment analysis was conducted to further explore the
   possible functions of the targets. In this section (see Additional file
   [119]3), Lipid and atherosclerosis (hsa05417), AGE-RAGE signaling
   pathway in diabetic (hsa04933), Kaposi sarcoma-associated herpesvirus
   infection (hsa05167), Hepatitis B (hsa05161), Fluid shear stress and
   atherosclerosis (hsa05418), Pertussis (hsa05133), Yersinia infection
   (hsa05135), Shigellosis (hsa05131), Chagas disease (hsa05142),
   Toll-like receptor signaling pathway (hsa04620), C-type lectin receptor
   signaling pathway (hsa04625), Th17 cell differentiation (hsa04659),
   Pancreatic cancer (hsa05212), Neurotrophin signaling pathway
   (hsa04722), Human cytomegalovirus infection (hsa05163), Coronavirus
   disease-COVID-19 (hsa05171), IL-17 signaling pathway (hsa04657),
   Endocrine resistance (hsa01522), Salmonella infection (hsa05132), T
   cell receptor signaling pathway (hsa04660), TNF signaling pathway
   (hsa04668) were significantly enriched (Fig. [120]2B). The KEGG pathway
   diagram disclosed that a series of target genes constituted the
   Toll-like receptor signaling pathway, including AKT, NFKB, ERK, JNK,
   IL6, and p38 MAPK (Fig. [121]2C). Therefore, it is reckoned that QHF
   exerts therapeutic effects through regulating immune-related functions.
   Moreover, the molecular docking verified a strong affinity of active
   components binding to targets in the predicted pathway (Fig. [122]2D).
   The docking parameters of each target and binding free energies were
   listed in Table [123]2. The results indicated that flavonoids of QHF,
   including quercetin, luteolin, kaempferol, and wogonin, had a strong
   affinity with core targets in terms of binding free energy.
   Interestingly, ASTX029, the novel inhibitor of ERK [[124]52], binds to
   ERK with a binding free energy of − 8.3, identical to that of
   quercetin, suggesting a high affinity of quercetin binding with ERK.
   The ligands and receptors were bound mainly by hydrogen bonds, which is
   positively related to the binding affinity. For example, luteolin could
   bind to TLR2, AKT, and JNK closely, with 6, 4, and 5 hydrogen bonds,
   respectively, and quercetin bound to ERK with 6 hydrogen bonds. Apart
   from the hydrogen bonds, the secondary effects were hydrophobic
   interaction of amino. These results further revealed the affinity of
   the active components of QHF binding to the transcriptionally
   dysregulated genes of B-cell lymphoma, which is the foundation of drugs
   to take effect.
Table 2.
   The molecular docking parameters
   Targets PDB ID Ligands Size of cube (X * Y * Z)
   (nm^3) Center grid box (X, Y, Z) Binding free energy (kcal/mol)
   TLR2 2Z80 Luteolin 100*100*100 −1.516 −8.3
   −14.704
   −14.946
   ERK 7AUV Quercetin 96*80*124 0.759 −8.3
   4.044
   37.087
   AKT 3O96 Luteolin 104*90*126 52.684 −9.9
   76.365
   34.530
   JNK 2NO3 Luteolin 104*90*126 15.902 −8.8
   58.495
   0.023
   [125]Open in a new tab
Antitumor effects of QHF in combination with ADM in vivo
   In this study, we first investigated the in vivo effects of QHF and
   Adriamycin (ADM) combination therapy on the growth of B cell lymphoma
   in A20 tumor-bearing mice. The study scheme is depicted in
   Fig. [126]3A. Mice bearing subcutaneous A20 tumors in QHF + ADM (n = 5)
   and ADM (n = 6) groups were compared with mice in the control group
   (n = 5). The tumor volume on day 12 was 744.07 mm^3 (QHF + ADM group),
   1275.23 mm^3(ADM group), and 1829.89 mm^3 (control group), respectively
   (Fig. [127]3B). Compared with the control group, the tumor volume of
   the QHF + ADM group was significantly lowered (vs. control group,
   p < 0.05). However, the ADM group showed no significant difference (vs.
   control group, p > 0.05), but exhibited a declining trend (Fig.
   [128]3C, D). There was a significant difference in TGI between
   QHF + ADM group and ADM group on day 12 (p < 0.01) (Fig. [129]3E).
Fig. 3.
   [130]Fig. 3
   [131]Open in a new tab
   QHF + ADM significantly inhibits the growth of A20 in the tumor-bearing
   mice model. A. Scheme of the animal experiment protocol. B. Photographs
   of all the tumor blocks collected from the Control group (n = 5), ADM
   group (n = 6), and QHF + ADM group (n = 5). C. Tumor volume of the
   tumor blocks collected from mice on day 12. It showed that QHF + ADM
   group (n = 5) significantly lowered tumor volume when compared with the
   control group (n = 5, p < 0.05). But there is no significance between
   the ADM group (n = 6, p > 0.05) and the control group (n = 5,
   p > 0.05). D. The tumor volume changes with time of the three groups.
   The tumor growth of the QHF + ADM group (n = 5) was the slowest of the
   three groups, followed by the ADM group (n = 5). E. Tumor growth
   inhibition rate (TGI, %) of the treatment groups. It showed that
   QHF + ADM (n = 5) could significantly inhibit tumor growth in an A20
   murine model (vs. ADM (n = 5), p < 0.01). Data are presented as the
   mean ± standard errors, *p < 0.05
QHF in combination with ADM increases tumor-infiltrating CD4+ and CD8+ T
cells
   In light of the systematic pharmacology research we did previously, we
   predicted that QHF probably exerts such tumor inhibition effect through
   modulating immune functions by activating the Toll-like receptor (TLR)
   signaling pathway. To verify the prediction, the tumors harvested were
   used to explore the differences in TME among the three groups. Sixteen
   tumor-bearing mice (three groups) were used, and their tumor blocks
   were stained for tumor-associate macrophages (TAMs) and
   tumor-infiltrating lymphocytes (TILs) via IHC. Chemokines and
   cytokines, including CXCL10, IL-2, IL-17, IL-6, IFN-γ, and TGF-β were
   also tested with ELISA. The IHC staining showed that there is no
   significance among the three groups in the proportion of M1 macrophages
   and M2 macrophages, but the combination group displayed a tendency to
   decline in the proportion of M2 macrophages (see Additional file
   [132]4). Interestingly, the proportions of CD4+ and CD8+ T-cells in the
   tumor tissues were significantly higher in the combination therapy
   group than in the ADM monotherapy group (Fig. [133]4A-D). As for the
   CD4+ T-cell ratio, the QHF + ADM group showed a significant difference
   when compared with the control group (p < 0.01) and the ADM monotherapy
   group (p < 0.01) (Fig. [134]4A, B). Besides, there was a significant
   increase of CD8+ T-cells in the combination group (vs. control group,
   p < 0.05; vs. ADM group, p < 0.05) (Fig. [135]4C, D). The ELISA
   revealed that mice in the combination group with QHF + ADM had higher
   levels of CXCL10 (v.s. Control, p < 0.05) and IL-6 (v.s. Control,
   p < 0.01). Meanwhile, the levels of TGF-β displayed a declining
   tendency, although the difference was not significant (Fig. [136]4E).
   Neither significant changes nor tendencies were observed for IL-2 and
   IL-17 (data not shown, see Additional file [137]5). The PET-CT images
   showed that compared with the pretreatment scans, the posttreatment
   mice had a relatively higher SUV[max] value (Fig. [138]4F). As for the
   mouse receiving ADM+ QHF, seven days after the therapy was
   discontinued, the volume of tumor enormously increased but the SUV[max]
   value lowered from 4.3 to 3, instead. These results suggested that
   combination therapy could remarkably enhance the level of
   tumor-infiltrating lymphocytes, especially the CD4+ and CD8+ T cells.
Fig. 4.
   [139]Fig. 4
   [140]Open in a new tab
   QHF + ADM recruited CD4+ and CD8+ T cells in the A20 tumor sites. A.
   Representative images of IHC staining for CD4+ T cell among the three
   groups (200×). B. Bar plot of the positive cell rate of CD4+ T cells of
   the control group (n = 3), ADM group (n = 3), and QHF + ADM group
   (n = 3). C. Representative images of IHC staining for CD8+ T cells
   among the three groups (200×). D. Bar plot of the positive cell rate of
   CD8+ T cells of the control group (n = 3), ADM group (n = 3), and
   QHF + ADM group (n = 3). E. Bar plot of TGF-β, IFN-γ, CXCL10, and IL-6
   levels based on ELISA of the control group (n = 3), ADM group (n = 3),
   and QHF + ADM group (n = 3).F. ^18F FDG PET-CT scan of the ADM group
   (n = 1), and QHF + ADM group (n = 1). G. Expression of TLR2, p38 MAPK,
   and β-actin by western blot. Data are presented as the mean ± standard
   errors, *p < 0.05; **p < 0.01
QHF exerts an antitumor effect by targeting the toll-like receptor signaling
pathway
   The protein levels of TLR2, p38 MAPK, and β-actin expression were
   detected by western blot in the present study. As shown in Fig.
   [141]4G, compared with the Control group (n = 3) and the ADM group
   (n = 3), the combination group (n = 3) significantly increased the
   expressions of TLR2 (v.s. Control group, p < 0.001; v.s. ADM group,
   p < 0.0001) and p38 MAPK (v.s. Control group, p < 0.001) (see
   Additional file [142]6).
Discussion
   B cell non-Hodgkin lymphoma is a highly heterogeneous neoplasm, and its
   most common subtype is diffuse large B cell lymphoma. Although it is
   curable to most patients, there are still 30-40% recurrence or
   refractory cases [[143]53]. And cancer immunotherapies are initially
   used in these settings. The TME, especially the immune landscape within
   it, is found to be crucial in the responses to the immunotherapies
   [[144]54]. The TME of B-cell lymphoma mainly consists of T cells,
   macrophages, natural killer (NK) cells, stromal cells, blood vessels,
   and extracellular matrix [[145]55, [146]56]. A combination therapy
   targeting TME has proven efficacy and displayed great potential for
   novel immunotherapies. As a traditional formula, QHF consists of four
   different herbs with various functions. In this study, we verified the
   tumoricidal effect of QHF in combination with ADM using a murine model
   and explored its underlying mechanisms with the guidance of systemic
   pharmacology.
   Systemic pharmacology is a comprehensive approach to clarify the active
   compounds and drug targets, as well as to predict the mechanisms of
   traditional Chinese medicinal formulas. Our work displayed that QHF has
   541 significant targets in B cell lymphoma, among which MAPK1/ERK2 and
   AKT1 are mostly enriched. Based on KEGG pathway enrichment analysis, we
   discovered that the TLR signaling pathway, which plays a critical role
   in activating both innate and adaptive immune responses, was one of the
   major pathways that QHF might trigger. In addition, we filtered out
   five active components of QHF, i.e., quercetin, luteolin, kaempferol,
   wogonin, and stigmasterol, that might be the most promising ingredients
   to represent QHF. The molecular docking technique also authenticated
   the ligation of the five components and targets.
   Our murine model built with an A20 cell line showed that compared with
   ADM monotherapy, the QHF + ADM group could significantly inhibit the
   tumor growth. Meanwhile, the tumor-infiltrating CD4+ and CD8+ T cells
   of the QHF + ADM group were remarkably increased. Research demonstrated
   that anti-CD20 mAb, such as rituximab, could achieve a long-lasting
   “vaccinal effect” with the presence of both CD4+ and CD8+ T cells
   [[147]57, [148]58], indicating the potential value of QHF as an add-on
   therapy to the R-based therapy for lymphoma patients. In this study,
   one mouse from each group was picked out to receive the ^18F-FDG PET-CT
   scan before and after the treatment, respectively. Notably, although
   the posttreatment tumor volume of the combination group is smaller than
   the ADM group, the SUV[max] of the combination group is higher. What’s
   more, after the treatment was discontinued for 1 week, the SUV[max] of
   the combination group decreased to a level lower than that of
   pretreatment. Evidence showed that SUV[max] is positively related to
   the TILs and TAMs [[149]59, [150]60]. And a provoking study published
   in Nature this year has proven that it is the myeloid cells, followed
   by T cells, that uptake most FDG rather than tumor cells [[151]61].
   Therefore, the dynamic changes of SUV[max] of the combination group
   might indicate the level changes of infiltrating T cells. The ^18F-FDG
   PET-CT scan provided an additional description of the increased
   infiltrating T cells in the QHF + ADM group.
   According to the immunophenotype of TME, tumors can be divided into
   “hot” tumors and “cold” tumors. Hot tumors present with rich TILs,
   whose initial immune response is obstructed by upregulation of immune
   checkpoints or immunosuppressive cells. Anti-PD-1/PD-L1 antibodies
   block tumor immune evasion by protecting CD8+ T cells from PD-1
   mediated cell death [[152]62, [153]63]. However, such ICI has limited
   efficacy in ‘cold’ tumors, where immune cells are less abundant or
   completely absent in TME [[154]64]. Mullins et al. [[155]65] found that
   TLR agonists tuned TME to inflamed immunophenotype by activating CD8+ T
   cells, thus enhancing the antitumor effect of ICIs. This has
   highlighted the promising value of TLR agonists. TLR is a big family
   consisting of TLR2, TLR4, TLR7, TLR8, and other receptors. TLR
   signaling pathway starts with TLR receptor and ligand recognition,
   followed by TLR dimerization and a signaling cascade to produce
   inflammatory molecules. In this study, we first predicted that the
   active components of QHF bind to TLR2 with hydrogen bonds via molecular
   docking methods. In theory, when the components of QHF bind to TLR2 and
   activate the TLR2 signaling pathway, the constitutive MAPK or NF-κB
   signaling pathway will be triggered to express various cytokines like
   IL-6, TNF-α, and IFNs. It is also reported that activation of TLR2 can
   reverse mouse Treg suppression, promote and generate efficient memory T
   cells by increasing IFN-γ level, and enhance the cytotoxic activity of
   CD8+ T cells [[156]66]. Besides, ADM can activate p38 to regulate the
   expression of topoisomerase II, inducing apoptosis of lymphoma cells.
   Therefore, QHF + ADM could enhance the antitumor effect via different
   mechanisms theoretically. In the present study, the IL-6 level was
   up-regulated in the QHF + ADM group, but the levels of TNF-α and IFN-γ
   were not remarkably changed. As the IL-6 production regulation is
   controlled through both transcriptional and posttranscriptional
   mechanisms [[157]67], it remains elusive whether the increased IL-6 is
   the result of TLR signaling pathway activation or not. But the increase
   in CD4+ and CD8+ T cells in TME, as well as up-regulated CXCL10 and
   TLR2 in the combination group, indicated QHF + ADM could modulate TME
   by targeting TLR2.
   As aforementioned, the immune cells within TME can interact with the
   tumor cells directly or through chemokines and cytokines to impact the
   behavior of the tumor and response rate [[158]68]. A variety of
   chemokines are expressed by immune cells, including CCR7, CXCL9,
   CXCL10, CCL2, and IFN-γ. Secreted by effector T cells, IFN-γ can induce
   the expression of CXCL10, which in return attracts even more T cells
   into the TME, thus forming a positive feedback loop [[159]69–[160]73].
   Despite IFN-γ, TLR agonists also elicit cytokines and chemokines
   expression in multiple cells [[161]65]. In a melanoma model, TLR2/6
   agonists + IFN-γ can induce CXCL10 production, leading to more T cells
   migrating into TME [[162]74]. In our study, the combination group
   displayed upregulated levels of TLR2 and CXCL10, indicating that QHF
   might target TLR signaling pathway to promote CXCL10 expression with
   the presence of IFN-γ, and consequently increased T cell infiltration
   and IFN-γ expression in return. Transforming growth receptor beta
   (TGF-β), expressed by both tumor cells and tumor-associated cells,
   directly inhibits T cell proliferation and activation while promoting
   the survival of immunosuppressive Tregs. Therefore, in the context of
   TME, upregulated TGF-β may lead to T cell dysfunction and limited
   efficacy of cancer immunotherapies [[163]75]. Moreover, with the
   presence of IL-6, TGF-β may also promote CD4+ T cell differentiation
   along a Th17 pathway, which was predicted in the KEGG enrichment
   analysis as mentioned before. Although IL-6 is associated with lymphoma
   proliferation and poor prognosis, it is reported that IL-6 can prevent
   apoptosis of T cells by upregulation of Bcl-2 and promote T cell
   proliferation via the TCR pathway [[164]76]. Also, evidence shows that
   increased IL-6 is associated with highly functional CAR T cells and
   their expansion [[165]77]. Consistent with these findings, we observed
   a downregulated level of TGF-β, as well as an upregulated level of IL-6
   in the combination group, suggesting that QHF may aid to restore T cell
   function and survival in the TME. Taken together, we speculated that
   QHF took effect through modulating the TIME via targeting the TLR
   signaling pathway.
Conclusion
   QHF in combination with ADM improves the anti-lymphoma effect of ADM by
   modulating TME. Besides, with a method of systematic pharmacology, the
   active components of QHF were identified. This research established a
   scientific method for analyzing chemical components and verifying
   predicted mechanisms of Chinese herbal medicine.
Supplementary Information
   [166]12906_2022_3660_MOESM1_ESM.pdf^ (74.3KB, pdf)
   Additional file 1: Figure S1. Pre-study data of the effective dosage of
   QHF. A.The image of the tumor blocks of the three groups.
   B.Thebodyweight of the mice in three groups during the experiment. C.
   The tumor blocks weight of the three groups. Compared with the LD group
   (n=5), the tumor weight was significantly decreased in the HD group
   (n=5, **p<0.01). Compared with the MD group (n=5), the tumor weight was
   remarkably lowered in the HD group (n=5, *p<0.05)
   [167]12906_2022_3660_MOESM2_ESM.xlsx^ (8.6KB, xlsx)
   Additional file 2: Table S1. Total input gene list
   [168]12906_2022_3660_MOESM3_ESM.xlsx^ (26.9KB, xlsx)
   Additional file 3: Table S2. KEGG pathway enrichment results
   [169]12906_2022_3660_MOESM4_ESM.pdf^ (379.4KB, pdf)
   Additional file 4: Figure S2. IHC stainingsfor M1 and M2 macrophages.
   A. Representative images of IHC staining for M1 macrophages among the
   three groups (200×). B. Bar plot of the positive cell rate of M1
   macrophages of the control group (n=3), ADM group (n=3), and QHF+ADM
   group (n=3). C. Representative images of IHC staining for M2
   macrophages among the three groups (200×). D. Bar plot of the positive
   cell rate of M2 macrophages of the control group (n=3), ADM group
   (n=3), and QHF+ADM group (n=3).Ns: p>0.05
   [170]12906_2022_3660_MOESM5_ESM.pdf^ (11.2KB, pdf)
   Additional file 5: Figure S3. ELISA results of IL-2 and IL-17. A.
   Concentration of IL-2 of the control group (n=3), ADM group (n=3), and
   ADM+QHF group (n=3). There is no significant difference between the
   three groups (p>0.05). B. Concentration of IL-17 of the control group
   (n=3), ADM group (n=3), and ADM+QHF group (n=3). There is no
   significant difference between the three groups (p>0.05)
   [171]12906_2022_3660_MOESM6_ESM.pdf^ (36.2KB, pdf)
   Additional file 6: Figure S4. Image J results of the WB bands. A. Bar
   plot of the expression of TLR2. Compared with the Control group (n=3)
   and the ADM group (n=3), the ADM+QHF group (n=3) significantly
   increased the expressions of TLR2 (v.s. Control group, ***p<0.001; v.s.
   ADM group, ****p<0.0001). B. Bar plot of the expression of p38.Compared
   with the Control group (n=3), the level of p38 MAPK was remarkably
   increased in ADM group (n=3, ***p<0.001) and ADM+QHF group (n=3,
   ***p<0.001)
Acknowledgments