Dendritic Cell Cancer Therapy Guide
Cancer treatment is changing fast, and one of the most talked-about ideas in recent years is teaching the immune system to do more of the fighting. Immunotherapy has already become a standard option for many cancers, and in some advanced stages, it’s helped patients live months or even years longer than older treatments alone.[1]
At the same time, global cancer cases keep rising - the WHO estimates nearly 20 million new cancer diagnoses every year worldwide.[2] Therefore, researchers are urgently seeking smarter, more targeted ways to leverage the body’s own defenses rather than relying solely on chemotherapy and radiation. That’s where dendritic cell therapy comes in: a still-evolving, highly personalized approach that is slowly moving from experimental labs and clinical trials into real-world clinics and patient conversations.[3][4]
This guide unpacks dendritic cells for cancer treatment in plain language. Reading this grounded, fact-based overview helps to navigate a dendritic cell-related topic, to make an informed decision on a further treatment choice.
Understanding Dendritic Cells & the Immune System
Before we get into how dendritic cell therapy works, it helps to zoom out and look at the bigger picture of your immune system. In this section, we'll explore how the body detects and fights malignant cells, and see the tricks cancer uses to slip past those defenses.
What Are Dendritic Cells?
To understand dendritic cell therapy, it helps to first picture the cells themselves as the 'event organizers' of your immune system. Dendritic cells are part of your immune system. Most of the time, they sit quietly in tissues like the skin, lungs, and gut, constantly sampling their surroundings. When they detect something dangerous, such as a virus, bacterium, or abnormal cancer cell, they grab tiny fragments and travel to the nearest lymph node. There, they present these fragments to other immune cells (mainly T-cells) and essentially say: "This is what the enemy looks like – go find anything that matches." Because of this role, dendritic cells are called antigen-presenting cells and act as key 'gatekeepers' between the outside world and an immune response.[5][6][7]

Dendritic cells themselves are not rare oddities. They are part of a much larger family of white blood cells that constantly circulate and patrol the body. In a single drop of blood, you can find millions of white blood cells, and a small but very important fraction of these can become dendritic cells under the right conditions.[5][7]
They were first recognized as a distinct cell type in the 1970s by Canadian immunologist Ralph Steinman.[8] This discovery eventually earned him a share of the Nobel Prize in Physiology or Medicine in 2011.[9] Because it helped explain how vaccines and immune responses initiate. Since then, researchers have identified several subtypes of dendritic cells, each with slightly different jobs. However, they all share the same core task: collecting information about potential threats and training the rest of the immune system to respond more effectively.
How the Immune System Fights Cancer
Your immune system is not only on the lookout for germs. It also keeps an eye on your own cells to make sure they’re behaving.[10] Every day, billions of cells in your body divide and copy their DNA. However, mistakes inevitably happen. Most of those errors are harmless or get fixed, but some can push a cell toward becoming cancerous.[11]
The immune system runs a quality-control process called “immune surveillance.” During this procedure, specialized cells look for anything that appears abnormal. T-cells, B-cells, natural killer (NK) cells, and dendritic cells all play a part in this quiet, constant patrol.[12]
Dendritic cells (DC) are the scouts in this process. They pick up suspicious molecules from damaged or abnormal cells and carry them to the lymph nodes. Here, a large number of T cells are waiting. There, dendritic cells display these molecules like “wanted posters.” If a T-cell’s receptor recognizes one of these posters as dangerous, that T-cell becomes activated. Then, they start multiplying and travel through the bloodstream back to the tissue where the problem started.[13] In successful cases, this leads to the destruction of emerging cancer cells before a tumor has the chance to grow big enough to be seen on a scan.[14]
Modern immunotherapy drugs can help people with advanced cancers live years longer.[15][16] That is why there is strong, real-world evidence that the immune system can be a potent anti-cancer force.
Why Cancer Sometimes Escapes the Immune System
Given all that, a natural question is: if the immune system is so vigilant, why does cancer ever manage to grow at all? One reason is timing. Cancer doesn’t usually appear overnight; it develops gradually, as cells accumulate multiple genetic changes over months or years.[17]
In the early stages, the differences between a pre-cancerous cell and a healthy one can be subtle. So the immune system may not recognize it as a serious threat.[18] When abnormal cells multiply, they are more likely to evolve tricks for hiding. Some tumors reduce the signals on their surfaces that would usually shout “I’m dangerous,” making it harder for T cells to spot them. Others release chemical signals that attract cells, which actually calm the immune response rather than activate it. Such masking turns the area around the tumor into a kind of “immunological fog” where defense cells become confused or exhausted.[19]
On top of that, the body itself has safety brakes designed to prevent the immune system from going too far and attacking healthy tissues. These brakes, known as immune checkpoints, are essential for preventing autoimmune diseases.[20] Nonetheless, cancers can learn to exploit them. By displaying specific molecules, a tumor can press the brakes on nearby T cells. Such a scenario tells them to stand down, even if those cells are capable of attacking it.[21]
The result is a paradox: you can have a strong immune system on paper, yet a tumor grows because it has found ways to stay below the radar.[22] Dendritic cell therapy aims to rebalance this ratio in favor of the immune system.[23] The treatment provides more explicit instructions and a sharper memory of what the cancer looks like.
What Is Dendritic Cell Cancer Therapy?
Dendritic cell cancer therapy is a type of personalized immunotherapy. The treatment aims to enhance your immune system's ability to recognize and attack tumor cells. Instead of attacking the tumor directly, doctors focus on the cells whose natural job is to show the immune system what to fight: dendritic cells.[24][25]
In this approach, immune cells are taken from the patient, guided in the lab to behave like active dendritic cells, exposed to cancer material, and then returned to the body. The goal is simple in principle, even if the science is complex: give your immune system a clearer, sharper picture of your specific tumor so T cells can more easily recognize and attack it.[26]
DC therapy is not just a lab fantasy. One dendritic cell–based product, Sipuleucel-T (Provenge), has been approved in the United States for certain men with advanced prostate cancer.[4] Other approvals come from India (APCEDEN), South Korea (CreaVax-RCC), and Brazil (HybriCell).[28][29][30]

Encouraged by good results, researchers have been testing different forms of dendritic cell therapy in various cancers. Beyond branded products, doctors worldwide use dendritic cells across different platforms by varying the source and delivery route to optimize T-cell priming against cancer.[31][32]
How It Differs from Other Immunotherapies
At first glance, dendritic cell therapy can seem similar to other modern immunotherapies. Still, it plays a different role in the immune “chain of command.”
- Immune Checkpoint Inhibitors. Checkpoint inhibitors, for example, are drugs that take the brakes off T cells that already recognize the tumor.[33]
- CAR-T Cell Therapy. Another well-known approach goes even further: T cells are extracted from the body, genetically engineered to recognize a specific target, and then infused back.[34]
Dendritic cell therapy operates a step earlier in the process. Instead of supercharging or rewriting the “soldiers” (T cells), it tries to improve the “intelligence officers” that brief them.[35] By giving dendritic cells better access to tumor material and encouraging them to present that information clearly, the treatment aims to generate a broader, more tailored T-cell response.[35][36]

Right now, this approach is usually seen as complementary to surgery, chemotherapy, radiation, or other immunotherapies rather than a complete replacement.[35]
Types of Dendritic Cell Therapy
Even within this single idea, there is not just one “dendritic cell vaccine,” but a family of related techniques.

Autologous: From Patient’s Blood
The most common version uses cells from the patient’s own blood. It is known as autologous dendritic cell therapy.[37] Doctors collect white blood cells, encourage some of them in the lab to become dendritic cells, and then expose those cells to information about the tumor.[38]
- Tumor Lysate-Loaded DCs. One popular way to do this is with tumor lysate. Lysate is essentially a “soup” made from pieces of the patient’s own tumor, usually obtained during surgery or a biopsy. Because this mixture contains many different proteins, including those generated by random mutations, it can provide dendritic cells with a comprehensive and personalized set of targets to present to the immune system. Such a DC type is beneficial in cancers like glioblastoma or ovarian cancer, where tumors often carry many unique genetic changes and have been challenging to treat with more narrowly focused strategies.[39][40]
- Peptide-Loaded DCs. Another strategy relies on peptide-loaded dendritic cells. The “lesson plan” is made from selected short fragments of proteins (peptides) known to be associated with a given cancer type or common mutation. Instead of feeding dendritic cells the whole tumor soup, researchers give them a curated menu of well-studied markers. Using peptides can standardize the manufacturing process. It may not require a large tumor sample, which is an advantage when getting tissue is risky or difficult. The trade-off is that this approach only covers the chosen targets; if a particular tumor doesn’t rely heavily on those proteins, or later evolves to lose them, the immune response may weaken.[41][42]
Allogeneic: From Donors
There is another scenario: using dendritic cells or precursors from healthy donors, so-called allogeneic products.[43][44] In theory, these “off-the-shelf” vaccines could be produced in larger batches and made available more quickly and cheaply, without tailoring each dose to one person.[25] However, they face additional hurdles, such as the risk of cell rejection or ineffective communication with each patient’s immune system, so most of the real-world experience to date has come from personalized, autologous vaccines.
Another Types
There are also other, more experimental types of dendritic cell–based approaches. Among them are vaccines that use DNA or RNA instructions instead of tumor material.[45] Also, strategies that aim to activate dendritic cells directly in the body, rather than removing cells and modifying them in the lab.[46] Because these methods are still at an early stage and not widely available in routine care, this guide focuses mainly on the better-studied, patient-specific (autologous) vaccines that readers are most likely to encounter in real-world clinics and clinical trials.[47]
Across all these variations, regardless of the type used, the core principle remains the same: dendritic cells give your immune system better, more detailed information about what to attack.[48]
Cancer Types Eligible for Dendritic Cell Therapy
When people first hear about dendritic cell therapy, a natural question is: “So which cancers can actually be treated with this?” At the moment, the picture of DC indications is quite uneven.[47] A small number of products have been formally approved in specific countries for defined cancer types.[35]
At the same time, a much longer list of cancers where dendritic cell vaccines are being tested in clinical trials. Plus, there is a grey zone in which some centers offer personalized vaccines on a case-by-case basis, even though regulators have not granted full approval.[49][50]
Fully Approved DC
The most widely known example is sipuleucel-T (Provenge), an autologous cellular immunotherapy approved by the U.S. Food and Drug Administration (FDA) in 2010.[51] It is used for men with asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. Provenge was the first therapeutic cancer vaccine of any kind to reach this level of regulatory recognition.[4]
Outside the United States, there are a few country-specific approvals that also fall into the dendritic cell category. In India, the Central Drugs Standard Control Organization (CDSCO) granted APCEDEN a commercial license in 2017.[52] It is an autologous dendritic cell–based product for use in several advanced solid tumors, including prostate, ovarian, colorectal cancer, and non-small cell lung cancer.[53]
In South Korea, CreaVax-RCC, there is an autologous dendritic cell vaccine targeting metastatic renal cell carcinoma.[54] The Korean regulator has authorized the drug as a therapeutic option for patients after kidney surgery.[30]
Taken together, approved options show that dendritic cell–based therapies have moved beyond the purely experimental stage.
Under Active Clinical Study
Beyond those few approved examples, dendritic cell vaccines are being studied across a broad spectrum of cancers. Mostly in early- and mid-stage clinical trials.

Solid Cancers
In solid tumors, some of the most active research has been in brain cancers, especially glioblastoma and other high-grade gliomas.[55] Vaccines are added to standard surgery, radiotherapy, and chemotherapy in an effort to delay recurrence in a disease that is typically aggressive.[56]
Melanoma has been another primary focus, both because it is historically responsive to immune-based treatments and because tumor tissue is relatively accessible.[57] Dendritic cell vaccines have been tested after surgery to reduce relapse risk and in metastatic disease alongside checkpoint inhibitors.[58]
Clinical trials have also explored dendritic cell approaches in non-small cell lung cancer and sarcomas (both in soft tissue and bone sarcomas).[59][60] It is sometimes combined with chemotherapy, radiation, or conventional immunotherapy.
In other solid cancers, dendritic cell immunotherapy shows prominent results. It can be offered for the following cancers: breast cancer[61], ovarian cancer[62], kidney cancer[63], bladder cancer[64], liver cancer[65], colorectal cancer[66], pancreatic cancer[67], head-and-neck cancers[25].
In many of these studies, patients receive the vaccine after standard treatment, when the visible tumor has been reduced. The goal was to mop up remaining cancer cells that are too small to detect on scans.
Blood Cancers
In the field of blood cancers, dendritic cell vaccines have been tested and used in the following diseases: acute myeloid leukemia (AML)[68], chronic myeloid leukemia (CML)[69], chronic lymphocytic leukemia (CLL)[70].
Additionally, it is applied to several types of non-Hodgkin lymphoma, Hodgkin lymphoma, multiple myeloma, and related bone marrow disorders.[71][72][73]
Often, the goal is to strengthen immune control in patients who are in remission but still at high risk of relapse. More indications come from patients who have minimal residual disease that conventional therapies have not eliminated.
Paediatric Oncology
Pediatric and young-adult cancers are also represented, though with smaller numbers. There are early-phase studies in pediatric brain tumors, neuroblastoma, Ewing sarcoma, and some other very high-risk or relapsed solid tumors where options are limited.[74][75]
Most indications are still in the stage where researchers are working out the best antigens, dosing schedules, and combinations, rather than offering a ready-made treatment that any hospital can provide.
Used in Some Scenarios
Finally, there is a more mixed category that sits between standard care and formal clinical research: individualized dendritic cell therapy offered by specific centers for selected patients. These uses are carried out in accordance with local hospital policies or compassionate-use frameworks.[76] In others, they operate in a less regulated, fully private environment.
Some private clinics, particularly in countries such as Germany, India, Japan, Czechia, and a few others, have developed in-house or partner programs that prepare personalized dendritic cell vaccines for a broad range of cancers. It spans from breast, lung, brain, and colorectal tumors to ovarian, prostate, and certain sarcomas or rare cancers. These programs may be offered when standard treatments have been exhausted, as part of “integrative” oncology packages, or occasionally earlier in the course of disease for patients who are highly motivated to explore every possible option.
Dendritic Cell Therapy Benefits & Risks
Dendritic cell (DC) therapy sits in an awkward middle ground: more than a lab curiosity, but not yet a universal standard. A few products are fully approved in specific settings; many more are in trials, and much of the “real-world” use occurs in small, highly selected patient groups. Here, we explore why it might be a good option, the exact results, and possible side effects.
Potential Benefits
Patients tend to compare treatments to decide which best fits them in a particular scenario. Now we focus on the benefits and advantages of a dendritic cell-based cancer vaccine.
- Personalized and Tumor-Specific. Most DC therapies are made from your own cells and often from your own tumor material. That means the vaccine can present a wide range of antigens that are genuinely present on your cancer, not just a single “generic” marker. In prostate and kidney cancer, a large meta-analysis of 906 patients found that when DC vaccines successfully triggered an immune response, the chance of any clinical benefit (tumor shrinkage or at least disease stabilization) rose to about 48–54%.[77]
- Long-Term Immune “Pressure”. Dendritic cell therapy often shows its value in how long people live, not in dramatic early scan responses. In the key trial of sipuleucel-T for metastatic castration-resistant prostate cancer, the vaccine improved 3-year survival from 23% to 32%. It extended median overall survival by 4.1 months.[78][79]
- Added on Top of Other Treatments. In many cancers, DC vaccines are used in combination with surgery, radiotherapy, chemotherapy, or other immunotherapies. For example, in adult glioblastoma, adding a tumor-lysate DC vaccine like DCVax-L to standard chemoradiotherapy has been associated with 2-year survival rates in the 30–60% range, compared with historical 2-year survival rates closer to 15–30% with standard treatment alone.[56][80]
- Generally Mild Toxicity. In many DC vaccine trials, the most common treatment-related problems are flu-like symptoms and injection-site reactions, usually grade 1–2 in severity. A broad review of DC vaccines concluded that they are “relatively safe” and that serious (grade 3–4) side effects occur in only a minority of patients.[56][81]
Dendritic Cell Therapy Effectiveness Summary
The success rate is one of the most crucial factors. Patients try to understand whether the DC vaccine will work for my case and what the likelihood is. This can be answered by reviewing similar cases and estimating a range for the possible response rate.

Because trials use different endpoints (tumor shrinkage, disease control, survival, or immune responses), it’s impossible to squeeze everything into one neat measure. The table below presents typical ranges reported in clinical studies for each cancer group, focusing on either objective response rate (ORR) or clinical benefit/disease control, where available.
| Cancer Type: Setting | Typical DC Effectiveness Range |
|---|---|
| Metastatic prostate cancer | Meta-analysis of 17 trials: ORR ~7–8%, but clinical benefit (CR+PR+SD) ~50–55%. Sipuleucel-T specifically improved median OS by 4.1 months (21.7 → 25.8 months; ~22–23% relative risk reduction in death).[77] |
| Metastatic renal cell carcinoma | Meta-analysis: ORR ~12–13%, clinical benefit ~45–50% in heavily pre-treated renal cancer patients.[77] |
| Advanced solid tumors | Phase II APCEDEN trial (51 patients): 28.9% response by RECIST, 42.1% by immune-related criteria, with a significant survival advantage vs best supportive care in a retrospective comparison.[83] |
| Glioblastoma | DCVax-L phase III and other series: 2-year overall survival rates of 30–50%, depending on MGMT and cohort, compared with typical historical 2-year survival rates of 15–30%; some smaller cohorts report 2-year OS of 52% with DC vaccination added to standard chemoradiotherapy.[56] |
| Melanoma | Reviews of DC vaccine trials report durable objective responses in roughly 5–15% of patients, with overall clinical response (including stable disease) in the 20–30% range; one TLPLDC melanoma study reported an ORR of 11% and 54% disease control.[84][85] |
| Non-small cell lung cancer | Small DC vaccine studies and overviews report ORR in the 20–40% range and disease control rates up to 60–75% when DC vaccines are combined with chemoradiotherapy or checkpoint inhibitors in selected stage III–IV patients.[86][87] |
| Ovarian cancer | Formal vaccine/adoptive cell therapy trials often show <10% objective response in heavily pre-treated patients, though clinic-level DC programs sometimes report 20–40% disease control and 45–65% immune response rates in selected series.[88] |
| Breast cancer | Early-phase DC and other vaccine trials usually show low ORR (often <10%) but high immune response rates in a much higher proportion of patients; broader therapeutic vaccine analyses across solid tumors report a pooled ORR of 13% and disease control of 39%, illustrating that only a minority achieve measurable shrinkage, but more achieve stabilization.[89] |
| Liver cancer | Systematic review of HCC vaccines (932 patients): overall pooled ORR 7%, but DC vaccines specifically 19% ORR (95% CI 11–29%), with median overall survival about 13.7 months and 1-year OS 40%.[90] |
| Colorectal & pancreatic cancers | Data for pure DC vaccines are limited; across therapeutic vaccine trials in KRAS-mutant CRC/pancreatic cancer, strong immune responses (e.g.,>80% KRAS-specific T-cell activation) are possible, but objective responses remain relatively rare, and most studies emphasize delayed relapse rather than high ORR.[91] |
| Head-and-neck cancers | Small DC vaccine or DC+checkpoint/chemo studies generally show disease control in 30–50% of patients, but accurate objective responses are less common, and numbers are too small to give a precise pooled ORR. (Most evidence is phase I/II, tens of patients per trial.)[92] |
| Acute myeloid leukemia | In post-remission AML, DC vaccination studies have reported measurable leukemia-specific immune responses and clinical responses in roughly 30–50% of evaluable patients, with some series showing 50% response rates in favorable-risk groups and suggesting prolonged relapse-free survival.[93] |
| CML, CLL, lymphomas, multiple myeloma | DC vaccines and DC–tumor fusion vaccines in these blood cancers have often induced immune responses and disease stabilization in a majority of patients (roughly 50–70%).[70][94] |
| Pediatric tumors | Early pediatric phase I/II trials primarily test safety. They report DC vaccination as feasible and well-tolerated, with immune responses and disease stabilization in about 20–40% of children, but low objective response rates and minimal patient numbers.[95][96] |
Possible Side Effects
While DC therapy is usually milder than chemotherapy or aggressive targeted drugs, it does have side effects. The profile depends on the product, but some patterns recur.

| DC Side Effect | Typical Frequency Range |
|---|---|
| Chills | Very common. In the sipuleucel-T registration trial, chills occurred in 51.2% of patients within 1 day of infusion. Smaller DC vaccine series reports chills or flu-like reactions in roughly 30–60% of patients.[98] |
| Fever (pyrexia) | In sipuleucel-T, fever was seen in 22.5% of patients within 24 hours of infusion. Many other DC trials report low-grade fevers in 20–40% of patients.[98][99] |
| Fatigue/tiredness | About 16% of patients in the sipuleucel-T group had fatigue within 1 day of infusion. Other DC vaccines show fatigue in roughly 15–30% of participants.[99] |
| Nausea | 14.2% in sipuleucel-T within 24 hours; similar low-to-moderate rates (often 10–20%) in other DC vaccine trials.[98] |
| Headache/flu-like feelings/muscle aches | Headache in 10–11% of sipuleucel-T patients; flu-like symptoms and myalgia were also reported more often than with placebo. Across DC studies, these “vaccine-type” symptoms are common but usually mild and short-lived.[100] |
| Injection-site reactions (redness, swelling, pain) | Very common in skin or subcutaneous DC injections, with many studies reporting local reactions in roughly 20–50% of patients. They are usually grade 1–2 and resolve on their own.[94][100] |
| Short-term grade ≥3 adverse events | In the sipuleucel-T trial, grade 3 or higher AEs within 1 day of infusion occurred in 6.8% of patients, compared with 1.8% in the placebo group; only 0.9% could not complete all three infusions because of infusion-related toxicity. In many other DC vaccine trials, serious vaccine-related toxicity is reported in <5–10% of patients.[98] |
| Autoimmune-type reactions, organ inflammation | Rare but theoretically possible. Large DC vaccine overviews and sipuleucel-T data do not show a strong signal for widespread severe autoimmune complications; cerebrovascular events in sipuleucel-T were 2.4% vs 1.8% with placebo and not statistically different.[98] |
Where It Fits in a Typical Treatment Journey
For most people, dendritic cell therapy is not the first thing their oncologist talks about. The starting point is still the traditional backbone of cancer care: surgery, radiotherapy, and drug treatments. Globally, experts estimate that around 80% of patients with cancer will need at least one surgical procedure during their illness.
Dendritic cell treatment sits atop this uneven foundation. It’s usually considered after standard options have been planned or used, or goes along with a more conventional solution.
Early-Stage vs Advanced Cancer
Where dendritic cell therapy might fit and what regimen to use depends a lot on when the cancer is caught. In many low- and middle-income countries, late diagnosis is a significant problem.
Cancer Stages 1-2
In early-stage cancers, especially when a tumor can be surgically obliterated, the main plan is usually straightforward: operate if possible, then consider radiotherapy or systemic drugs to reduce the risk of relapse.[101][102] Stages I or II cancer are where many dendritic cell vaccines are currently being studied. In brain tumors, kidney cancer, melanoma, and a few other cancers, researchers have tested these vaccines after surgery and standard radiochemotherapy.[103][104] DC is applied for patients whose visible disease has been reduced as far as possible. The idea is that when scans show little or no tumor, a dendritic cell vaccine might help the immune system patrol for leftover cancer cells that are too small to see.[68]
Cancer Stages 3-4
In advanced or metastatic cancer, treatment goals shift toward controlling growth and maintaining quality of life.[105] Drug therapies and radiotherapy take centre stage, and doctors may add typical immunotherapies when guidelines support them. Dendritic cell therapy, when considered, is usually scheduled once some control has been achieved (for example, when scans show partial remission or stable disease).[106] Then, DC can be used when standard treatment options have been largely exhausted, and the team is exploring clinical trials or individualized approaches.[107]
Summary of DC Treatment Scenarios & Logic
To make all of this more concrete, the table below shows where dendritic cell (DC) therapy is currently used or being studied in real-world settings. You understand why doctors might choose that timing, with a few examples and numbers to give a sense of scale. It’s not a rulebook, but a map of the most common scenarios you’ll see in trials and specialized centers.

| Timing | How DC Therapy Is Used: Examples & Logic |
|---|---|
| Before Primary Treatment | In some trials of newly diagnosed glioblastoma, patients are enrolled before starting standard chemoradiotherapy. Tumor tissue and blood can be collected, and a personalized DC vaccine can be prepared in parallel with the first phase of treatment. In one phase II study of a personal DC vaccine (DC-ATA), 57 patients enrolled, and 69% received all eight planned doses over about 6 months. That demonstrates that “planning” is logistically feasible in selected clinics.[108][109] |
| After Surgery | The tumor is first removed as completely as possible, and the vaccine is then given as adjuvant (add-on) therapy to help the immune system clear remaining cells. It is one of the most common DC vaccine scenarios. In glioblastoma, for example, a phase III study of the DCVax-L vaccine enrolled 331 patients after surgery and chemoradiotherapy. The analyses now pool data from thousands of patients across multiple trials using this “post-surgery vaccine” strategy. Similar post-surgery adjuvant DC approaches are being tested in melanoma and other solid tumors.[110][111] |
| After Chemo-Radiotherapy | In high-grade brain tumors, DC vaccination is often slotted between the end of combined radiochemotherapy and the start of maintenance chemotherapy, or alongside the early maintenance cycles. Reviews of DC vaccination in glioblastoma describe schedules in which patients receive multiple injections every 2–4 weeks during the “post-radiochemotherapy window.” By 2023, at least 77 studies of DC vaccination in malignant glioma had been published, many of which used this timing.[112][113] |
| Along with Radiotherapy | Radiotherapy is used at some point in the treatment of roughly 50–60% of all cancer patients. There is growing interest in harnessing the immune-stimulating effects of radiation. Early DC-based strategies in head-and-neck and brain cancers often give the vaccine close to radiotherapy. This timing aims to turn radiation-damaged tumor cells into a stronger antigen source. These are usually small phase I/II studies with tens of patients each, designed mainly to test safety and biological signals.[114][115][116] |
| Along with Chemotherapy | One of the best-described combinations is chemotherapy + DC vaccine + cytokine-induced killer (CIK) cells in colorectal cancer. A meta-analysis pooled data from 871 patients across four clinical trials comparing chemotherapy alone with chemotherapy plus DC/CIK. The combination was feasible, and the immunotherapy component typically delivered around 10 million DCs and over a billion CIK cells per course, without introducing new severe toxicities. Similar chemo–DC combinations have been reported in breast, lung, liver, and kidney cancers in smaller cohorts.[117][118][119] |
| Along with Immunotherapy | Checkpoint inhibitors such as pembrolizumab, nivolumab, and atezolizumab are now standard options in several cancers. The drugs are typical for melanoma, lung, bladder, head-and-neck cancers, and some lymphomas. DC vaccines are being tested on top of this in early-phase studies, especially in melanoma and lung cancer. The logic is that the vaccine “shows” more tumor antigens while the checkpoint inhibitor keeps T cells active. Reviews describe this as a promising but still exploratory area. The trial sizes so far have usually been in the dozens rather than the hundreds.[120][121] |
| As Maintenance after Immunotherapy | In some glioblastoma protocols, patients receive a DC vaccine for many months as maintenance therapy. It's done alongside oral chemotherapy after the initial radiochemotherapy is complete. In the DC-ATA phase II trial, for instance, eight vaccine injections were planned over about 6 months. Most patients who started the program were able to stay on it long enough to receive all doses. The data suggest that prolonged "immune maintenance" schedules are workable in real-world glioma patients who are fit enough.[109] |
| Combined with Targeted Therapy | Targeted drugs such as sorafenib, regorafenib, and lenvatinib are a mainstay in advanced hepatocellular carcinoma (HCC). Because these agents rarely cure the disease on their own, reviews of HCC immunotherapy discuss DC vaccines as potential combination partners. It is especially relevant in patients whose tumors are controlled but not eliminated by TKIs. So far, these trials are small and early. Still, they reflect an essential principle: using DC therapy to deepen or prolong responses achieved with targeted drugs.[122][123] |
| Merged with Local Ablative Procedures | Local liver ablations like radiofrequency ablation (RFA) or microwave ablation are widely used for primary tumors and metastases when surgery is not possible. In HCC, for example, a single clinical trial combining RFA with a tumor-specific peptide vaccine reported better 1-year recurrence outcomes than RFA alone. More recent work has looked at pairing ablation with DC/CIK-based cellular immunotherapy. Such strategies typically involve dozens of patients per study. They are positioned as ways to "mop up" microscopic disease after the visible tumor has been burned or frozen.[124][125] |
| Instead of Conventional Systemic Therapy | The clearest example here is sipuleucel-T for prostate cancer. Label information and clinical policy documents describe a standard course of three infusions given at about 2-week intervals. Each is preceded by a leukapheresis procedure a few days earlier, so the whole program is usually completed in around 4–6 weeks. In practice, sipuleucel-T is often offered instead of or before chemotherapy in men who have relatively few symptoms. The DC here offers an immune-based option that doesn't add typical chemo-type side effects.[126] |
| Salvage Approach | Many DC trials focus on patients whose cancers have come back or failed standard treatment. In malignant glioma, for instance, a 2023 review counted 77 studies of DC vaccination, many in recurrent disease and mostly phase I or II, with typical trial sizes of 10–50 patients. In blood cancers, early AML studies have tested DC vaccines as salvage or relapse-setting therapies in tiny groups. One classic phase I/II trial enrolled 10 AML patients to explore immune effects and feasibility rather than to replace established chemotherapy.[113][127][128] |
| Intratumoral “in situ vaccination” strategy | Instead of giving DCs under the skin or into lymph nodes, some protocols inject them directly into the tumor, or use radiation and immune-stimulating agents to turn the tumor itself into a kind of vaccine factory. In glioma and head-and-neck cancer, for example, pilot studies have combined debulking surgery, chemoradiation, and intratumoral DC injections in small cohorts, often fewer than 30 patients per study, to test whether "in situ vaccination" can delay regrowth.[129] |
| Maintenance–Consolidation after Good Response | In acute myeloid leukemia, DC vaccination is being explored specifically as a post-remission consolidation strategy. Patients first receive standard chemotherapy to achieve remission. A DC vaccine course is added to help keep the disease under control. A phase I/II program in Norway followed 20 AML patients for 24 months of DC vaccination and monitoring. The data demonstrate that long-term DC vaccine schedules can be delivered safely in this setting. A recent systematic review identified 11 clinical trials of DC vaccination in AML overall.[68][131] |
| Secondary Prevention in High-Risk or Premalignant States | A more experimental use is "secondary prevention". Prevention here means treating people who do not have bulky, active cancer but have a very high risk of relapse or progression based on their history or lab markers. In AML and myelodysplastic syndromes, some DC vaccine strategies specifically target patients in complete remission but with high relapse risk, aiming to delay or prevent the return of leukemia. Reviews of these approaches highlight that individual trials are small (often 10–40 patients).[43][128][132] |
Compared to Standard Treatments
When comparing dendritic cell (DC) therapy to surgery, chemotherapy, or radiation, it’s crucial to understand that each is a distinct solution. Surgery and radiation focus on removing or destroying tumors. Chemotherapy spreads through the body to attack cancer cells directly. In contrast, DC therapy acts as a tool to educate the immune system. It doesn’t cut, burn, or poison the tumors. Instead, it trains the natural resistance to function more effectively over time.[24][26]
Because DC vaccines are mostly tested in small, early-phase studies, often after standard treatment or in selected, relatively fit patients, direct head-to-head comparisons with large, guideline-defining trials of chemo or radiotherapy are not really fair or statistically meaningful.
That said, there are some practical paths in which DC therapy differs from standard options. Drug treatments such as chemotherapy or a surgical approach have more risks. By comparison, dendritic cell-based vaccines have most often caused milder side effects. More serious DC treatment-related complications are reported much less frequently.[133]
Dendritic cell treatment is also highly personalized and is intended to leave behind an immune “memory.”[134] On the contrary, a single course of radiotherapy or a block of chemotherapy does not aim to do so.
Who Might Not Be a Good Candidate?
Dendritic cell therapy uses a patient’s own immune system. As a result, doctors often set strict selection criteria. Many early studies of dendritic cell vaccines included patients with good performance status, classified as ECOG 0 or 1.[135] This means they were fully active or had minor limitations in heavy physical work. Patients also needed stable organ function and no uncontrolled infections or autoimmune diseases. Consequently, many real-world patients, especially older adults with multiple health issues, are often excluded from these trials.
People with severely weakened immunity, such as those on high-dose steroids or immunosuppressants, may not be considered good candidates. The reason is that their bodies may not respond effectively to the vaccine.[136]
Patients with cancers that are growing really quickly might not be the best fit for vaccine treatment.[112] If it takes a week or longer to make the vaccine and then several more weeks for a complete set of injections, the tumor could end up getting a lot bigger during that time. Because of this, oncologists usually focus on treatments that can be started within a few days and that work right away to reduce the tumor size.
None of this means that dendritic cell therapy is automatically “better” or “worse” than other approaches. It simply reflects where it fits in the real-world treatment journey today. Despite the mentioned contraindications and limitations, it is reasonable to consult with dendritic cell clinics regarding your particular case. The actual application can vary widely depending on location and a provider's specific experience.
Step-by-Step: From Production to Administration
Dendritic cell therapy can sound complicated from the outside. Still, behind the scenes, it follows a fairly predictable path: collect your cells, teach them about the tumor, and then return them so your immune system can actually use that information. The exact details vary from clinic to clinic, but the main stages are surprisingly similar across most programs.

From Blood Draw to Lab: Collecting Cells
Everything starts with getting hold of the right immune cells. In many protocols, this is done with a procedure called leukapheresis. You sit or lie in a chair while blood is drawn from one arm, passed through a machine that filters out specific white blood cells, and then returned to your other arm. The entire circuit is closed, so you’re not “losing” liters of blood; the machine separates the cells it needs and keeps them.[137]
A typical leukapheresis session lasts 2–4 hours, depending on your veins, your weight, and the machine being used.[138] You’re usually awake the whole time, maybe reading or on your phone. Some people feel a bit cold or tired, or notice tingling from temporary changes in calcium levels, but serious complications are rare.[139]
In some centers, instead of full leukapheresis, a simple large-volume blood draw is enough, especially in smaller trials.[75] Once the cells are collected, they’re rushed to a specialized lab.
Turning Cells into Dendritic Cells
In your bloodstream, the cells that can become dendritic cells are mostly monocytes, a type of white blood cell. In the lab, technicians separate these monocytes from the rest of the collection and place them in sterile culture flasks containing carefully selected nutrients and signaling molecules. Over several days (often 5 to 7), sometimes a bit longer, these signals gently push monocytes to mature into dendritic cells.[140]
This section takes place in a clean, controlled environment that resembles a high-tech kitchen. The facility has filtered air, specialized cabinets, and strict contamination protocols. Staff members regularly check the cultures to ensure proper cell growth.[141] Before moving forward, the lab conducts quality control tests. These tests confirm that the cells look and behave like dendritic cells and that no harmful bacteria or fungi are present. Only after these checks are the cells ready to interact with the tumor material.
Creating the Personalized “Vaccine”
Once the dendritic cells are ready, they need to be introduced to the cancer's “face.” This is where the personalization really happens. If tumor tissue has been collected during surgery or biopsy, it can be processed into a tumor lysate. It is basically a finely blended mixture of proteins from that specific tumor.[143] The dendritic cells are incubated with this material so they can pick up and process the cancer proteins.
In other protocols, instead of whole-tumor lysate, the lab uses selected protein fragments (peptides) known to appear in certain cancers, or even strands of RNA/DNA that instruct dendritic cells to make those proteins themselves for a brief period.[142]
The end goal is the same: to load the dendritic cells with enough tumor-related information that, when they go back into your body, they know exactly what to show the immune system.
The finished product is usually prepared as multiple doses from a single collection. Some centers freeze individual portions in liquid nitrogen so they can thaw and inject them over several weeks or months without needing to repeat the entire lab process each time.[144]
How the Vaccine Is Given
When people hear “vaccine,” they often think of a quick jab in the upper arm, and in many dendritic cell protocols, that’s not far from the truth. The most common routes are intradermal (into the skin) or subcutaneous (just under the skin), sometimes in the upper arm, thigh, or abdomen.[145] In some studies, doctors inject dendritic cells directly into lymph nodes under ultrasound guidance or give them as a vein infusion.[146] The principle is the same: get them to the places where immune cells gather.
A typical schedule involves a series of injections, often every 1–3 weeks at the beginning, then more spaced out once the course is underway. Many treatment plans include 2-8 doses, sometimes more in longer maintenance programs.[97] Before each dose, you may have a quick check-up and basic blood tests to make sure it’s safe to proceed.
What Happens After Injection
After injection, the dendritic cells don’t just sit there; they start migrating.[82] Their job is to travel to lymph nodes, the small immune hubs dotted around your body, and display bits of the cancer proteins they’ve been loaded with. Inside those nodes, they meet T cells and other immune cells, and a kind of microscopic briefing takes place: “This is what we’re looking for. If you see anything with this pattern, attack.”[27]
If all goes well, some of those T cells become activated and start multiplying. They circulate through the blood and tissues, looking for cancer cells that carry the same markers they’ve been shown to hold. This is not an instant process. It can take weeks for a strong immune response to build up and longer for that response to translate into visible changes on scans.[27]
Long-Term Outlook
After the vaccination course finishes, follow-up usually resembles standard cancer care: regular visits, imaging (such as CT or MRI scans), and blood tests over months and years.[104] Some patients will show clear signs of ongoing immune activity against the tumor long after the last injection, while others may not. Because dendritic cell therapy is still evolving, many programs keep detailed, long-term records of their patients to understand who benefits most and how durable those benefits are.
For now, it’s best to see the whole process as a long game rather than a quick fix. The aim is not only to help the immune system react in the short term, but also to encourage a kind of immune “memory” that might keep watching for cancer cells over time.[130] Ideally, reducing the chances of the disease flaring up again, even if the visible part of the treatment ended months earlier.
Dendritic Cell Therapy Availability & Cost
Dendritic cell (DC) therapy is highly personalized and still relatively niche. Therefore, prices vary widely across countries, clinics, and treatment protocols. Most programs are paid out of pocket. The table below provides very approximate ranges for orientation, not as formal quotes.
DC Asses & Cost per Regions
| Country / Region | Typical cost range | Capacity |
|---|---|---|
| United States | $80,000–200,000 | Limited slots |
| United Kingdom | £40,000–80,000 | Limited slots |
| Germany | €19,000–50,000 | Largely assessed |
| India | $8,000–15,000 | Largely assessed |
| Japan | $17,000–45,000 | Limited slots |
| Hong Kong | $55,000–90,000 | Limited slots |
| Turkey | $10,000–28,000 | Largely assessed |
| Mexico | $35,000–110,000 | Largely assessed |
| Czech Republic | €20,000–30,000 | Limited slots |
| Canada | $45,000–80,000 | Occasional Clinical Trials |
| Ireland | €40,000–70,000 | Almost Unavailable |
| Netherlands | €35,000–60,000 | Occasional Clinical Trials |
| Denmark | €32,000–65,000 | Occasional Clinical Trials |
| Spain | €40,000–55,000 | Limited slots |
| Africa | $8,000–45,000 | Almost Unavailable |
| Australia | $40,000–95,000 | Occasional Clinical Trials |
| Israel | $43,000–85,000 | Limited slots |
Warning About Unproven or “Miracle Cure” Offers
Be very cautious of any clinic that advertises dendritic cell therapy as a “100% cure,” “no side effects,” or “works for all cancers”. Mainly, if they rely on testimonials rather than real clinical data, it's a classic red flag for unproven or unsafe offers.
To reduce your risk, always double-check where you’re going and who is treating you. Airomedical can help by listing vetted clinics & doctors, with verified profiles, credentials, and reviews. Here, patients are not relying only on glossy marketing when making serious decisions about their care.
Safety Note & Disclaimer
This guide is for general information only and is not a substitute for professional medical advice, diagnosis, or treatment. Cancer care is complex and highly individual: never start, stop, or change a therapy based solely on what you read online. Always discuss your options with a qualified oncologist or specialist who knows your specific case.
Dendritic cell therapies should be received only at properly regulated, reputable medical centers. Before making decisions, it’s essential to verify a clinic’s accreditation and a doctor’s training, experience, and track record. The Airomedical platform can help by showing verified clinic profiles and doctor credentials, along with genuine patient reviews. Here you can double-check that the team you’re considering is legitimate and appropriately qualified.
FAQ
How long does it take to see results?
With dendritic cell therapy, any benefit is usually measured in months, not days. Scans and blood tests are typically checked on the same schedule as with other cancer treatments (for example, every 2–3 months). Doctors look more at long-term control or delayed progression than at an immediate shrinking of the tumor.
Can I stop chemotherapy if I start dendritic cell therapy?
You should never stop chemotherapy or any standard treatment on your own because you’re starting a vaccine or immunotherapy. In almost all real-world protocols, dendritic cell therapy is used together with or after standard treatments, not as a replacement.
Is it safe for older patients?
Age alone doesn’t automatically rule someone out. Many trials have included patients in their 60s and 70s. Doctors focus more on overall health, organ function, and the robustness of the immune system than on the birthdate. An older person who is reasonably fit may be a better candidate than a younger person who is very frail.
Can I use dendritic cell therapy instead of surgery?
For tumors that can be safely removed, surgery is still one of the most effective tools to improve survival. Dendritic cell therapy is not designed to replace an operation that could remove the bulk of the cancer. It’s usually considered after surgery or when surgery isn’t possible, as an additional way to support long-term control.
Are there dietary supplements that boost dendritic cell therapy?
There’s no solid evidence that any over-the-counter supplement can specifically “boost” dendritic cell therapy. Some products can actually interfere with treatment or other medicines. It’s safest to focus on a balanced diet and discuss any vitamins or supplements with your oncologist before taking them.
What should I watch out for in medical offers?
Be cautious of clinics that promise guaranteed cures. Use only glowing testimonials instead of real data, or can’t clearly explain their training and regulatory status. Legitimate centers can demonstrate formal approvals, ethics oversight, and transparency about risks, limits, and the fact that results vary between patients.
Is dendritic cell therapy approved for my type of cancer?
At the moment, full regulatory approval exists only for a few specific situations in certain countries. In contrast, most other uses are still in clinical trials or offered in specialized programs. Whether it’s a realistic option for your own cancer depends on your diagnosis, where you live, and what trials or licensed products are accessible.
References
- Zhao, J., Graetz, I., Howard, D., Yabroff, K. R., & Lipscomb, J. (2025, July 7). Immune Checkpoint Inhibitors and Survival Disparities by Health Insurance Coverage Among Patients With Metastatic Cancer. JAMA Network Open, 8(7), e2519274. https://doi.org/10.1001/jamanetworkopen.2025.19274. Retrieved November 2025.
- International Agency for Research on Cancer (IARC). (2020, December 15). Latest global cancer data: Cancer burden rises to 19.3 million new cases and 10.0 million cancer deaths in 2020. Retrieved October 2025.
- Fan, T., Zhang, M., Yang, J., Zhu, Z., Cao, W., & Dong, C. (2023, December 13). Therapeutic cancer vaccines: advancements, challenges and prospects. Signal Transduction and Targeted Therapy, 8, 450. https://doi.org/10.1038/s41392-023-01674-3. Retrieved November 2025.
- Cheever, M. A., & Higano, C. S. (2011, June 1). PROVENGE (Sipuleucel-T) in Prostate Cancer: The First FDA-Approved Therapeutic Cancer Vaccine. Clinical Cancer Research, 17(11), 3520–3526. https://doi.org/10.1158/1078-0432.CCR-10-3126. Retrieved October 2025.
- Hicks, M. A., & Tyagi, A. (2023, May 1). Magnesium Sulfate. In StatPearls [Internet]. StatPearls Publishing. Retrieved October 2025.
- Nature Reviews Immunology. (2011, June 24). In Brief: The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Nature Reviews Immunology, 11, 439. https://doi.org/10.1038/nri3023. Retrieved November 2025.
- Aristizábal, B., & González, Á. (2013, July 18). Innate immune system. In Autoimmunity: From Bench to Bedside [Internet]. El Rosario University Press. Retrieved November 2025.
- NobelPrize. (2011). Ralph M. Steinman – Facts. Retrieved October 2025.
- NobelPrize. (2011, October 3). The Nobel Prize in Physiology or Medicine 2011 – Press release. Retrieved October 2025.
- Dunn, G. P., Old, L. J., & Schreiber, R. D. (2004, August). The Immunobiology of Cancer Immunosurveillance and Immunoediting. Immunity, 21(2), 137–148. https://doi.org/10.1016/j.immuni.2004.07.017. Retrieved November 2025.
- Pray, L. A. (2008). DNA Replication and Causes of Mutation. Nature Education, 1(1), 214. Retrieved November 2025.
- Ostrand-Rosenberg, S. (2008, February). Immune surveillance: A balance between protumor and antitumor immunity. Current Opinion in Genetics & Development, 18(1), 11–18. https://doi.org/10.1016/j.gde.2007.12.007. Retrieved November 2025.
- Xiao, Z., Wang, J., Yang, J., Guo, F., Zhang, L., & Zhang, L. (2025, April 24). Dendritic cells instruct T cell anti-tumor immunity and immunotherapy response. The Innovation Medicine, 3(2), 100128. https://doi.org/10.59717/j.xinn-med.2025.100128. Retrieved November 2025.
- Mlecnik, B., Bindea, G., Pagès, F., & Galon, J. (2011, January 21). Tumor immunosurveillance in human cancers. Cancer and Metastasis Reviews, 30, 5–12. https://doi.org/10.1007/s10555-011-9270-7. Retrieved October 2025.
- National Cancer Institute (NCI). (2019, September 24). Immunotherapy to Treat Cancer. Retrieved November 2025.
- American Cancer Society. (2025, August 7). What Is Immunotherapy?. Retrieved November 2025.
- International Agency for Research on Cancer (IARC). (2003). Mechanisms of tumour development (World Cancer Report 2003, Chapter 3). Retrieved November 2025.
- Tufail, M., Jiang, C.-H., & Li, N. (2025, July 31). Immune evasion in cancer: Mechanisms and cutting-edge therapeutic approaches. Signal Transduction and Targeted Therapy, 10, Article 227. https://doi.org/10.1038/s41392-025-02280-1. Retrieved October 2025.
- Li, M. (2025, October). Tumor Immune Evasion Mechanisms and Immunotherapy Strategies. Theoretical and Natural Science, 141(1), 109–115. https://doi.org/10.54254/2753-8818/2025.AU27725. Retrieved November 2025.
- ScienceDirect Topics. Immune Checkpoints – an overview. Retrieved October 2025.
- Buchbinder, E. I., & Desai, A. (2016, February). CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. American Journal of Clinical Oncology, 39(1), 98–106. https://doi.org/10.1097/COC.0000000000000239. Retrieved October 2025.
- Hossain, M. A. (2024). A comprehensive review of immune checkpoint inhibitors for cancer treatment. International Immunopharmacology, 143, 113365. https://doi.org/10.1016/j.intimp.2024.113365. Retrieved November 2025.
- Maeng, H. M., Olkhanud, P. B., Black, M., Highfill, S. L., Stroncek, D. F., & Berzofsky, J. A. (2025). Dendritic Cell Cancer Vaccines: A Focused Review. In Cancer Vaccines (Methods in Molecular Biology, Vol. 2926, pp. 51–56). Springer. https://doi.org/10.1007/978-1-0716-4542-0_4. Retrieved November 2025.
- Janikashvili, N., Larmonier, N., & Katsanis, E. (2010, January). Personalized dendritic cell-based tumor immunotherapy. Immunotherapy, 2(1), 57–68. https://doi.org/10.2217/imt.09.78. Retrieved November 2025.
- Mehrani, Y., Morovati, S., Keivan, F., Sarmadi, S., Shojaei, S., Forouzanpour, D., Bridle, B. W., & Karimi, K. (2025, May 30). Dendritic Cell-Based Cancer Vaccines: The Impact of Modulating Innate Lymphoid Cells on Anti-Tumor Efficacy. Cells, 14(11), 812. https://doi.org/10.3390/cells14110812. Retrieved November 2025.
- Caro, A. A., Deschoemaeker, S., Allonsius, L., Coosemans, A., & Laoui, D. (2022, August 21). Dendritic Cell Vaccines: A Promising Approach in the Fight against Ovarian Cancer. Cancers, 14(16), 4037. https://doi.org/10.3390/cancers14164037. Retrieved November 2025.
- Montgomery, L., & Larbi, A. (2025, April 16). Monitoring immune responses to vaccination: A focus on single-cell analysis and associated challenges. Vaccines, 13(4), 420. https://doi.org/10.3390/vaccines13040420. Retrieved November 2025.
- Najafi, S., & Mortezaee, K. (2023, August). Advances in dendritic cell vaccination therapy of cancer. Biomedicine & Pharmacotherapy, 164, 114954. https://doi.org/10.1016/j.biopha.2023.114954. Retrieved November 2025.
- PharmaBoardroom. (2019, January 21). Kyoung-June Lee – CEO, JW CreaGene, South Korea. Retrieved November 2025.
- Janes, M. E., Gottlieb, A. P., Park, K. S., Zhao, Z., & Mitragotri, S. (2024). Cancer vaccines in the clinic. Bioengineering & Translational Medicine, 9(1), e10588. https://doi.org/10.1002/btm2.10588. Retrieved November 2025.
- Clayton, G., Toffoli, E. C., & de Gruijl, T. D. (2025, November 18). Dendritic cell immunotherapy advances for solid tumors: Vaccination and modulation. Cell Reports Medicine, 6(11), 102412. https://doi.org/10.1016/j.xcrm.2025.102412. Retrieved October 2025.
- Cell Reports Medicine. Cell Reports Medicine, Volume 6, Issue 11. Cell Reports Medicine, 6(11). Retrieved November 2025.
- Al-Oudah, G. A., Alfalluji, W. L., & Al-Ajrash, A. M. N. (2025, June 30). Checkpoint Inhibitors in Cancer Immunotherapy: Mechanisms of Action and Resistance. Journal of Biomedicine and Biochemistry, 4(2), 124–149. https://doi.org/10.57238/jbb.2025.7432.1143. Retrieved November 2025.
- National Cancer Institute. (2025, February 26). CAR T Cells: Engineering Patients’ Immune Cells to Treat Their Cancers. Retrieved November 2025.
- Sheykhhasan, M., Ahmadieh-Yazdi, A., Heidari, R., Chamanara, M., Akbari, M., Poondla, N., … Kalhor, N. (2025, March). Revolutionizing cancer treatment: The power of dendritic cell-based vaccines in immunotherapy. Biomedicine & Pharmacotherapy, 184, 117858. https://doi.org/10.1016/j.biopha.2025.117858. Retrieved November 2025.
- Zanotta, S., Galati, D., De Filippi, R., & Pinto, A. (2024). Enhancing Dendritic Cell Cancer Vaccination: The Synergy of Immune Checkpoint Inhibitors in Combined Therapies. International Journal of Molecular Sciences, 25(14), 7509. https://doi.org/10.3390/ijms25147509. Retrieved November 2025.
- Kim, M. E., & Lee, J. S. (2025). Dendritic Cell Immunotherapy for Solid Tumors: Advances in Translational Research and Clinical Application. Current Issues in Molecular Biology, 47(10), 806. https://doi.org/10.3390/cimb47100806. Retrieved November 2025.
- da Silva Santos Duarte, A., Basso Zangirolami, A., Santos, I., Niemann, F. S., Honma, H. N., Amaro, E. C., … Olalla Saad, S. T. (2024, January–March). Production of dendritic cell vaccines using different methods with equivalent results: Implications for emerging centers. Hematology, Transfusion and Cell Therapy, 46(1), 30–35. https://doi.org/10.1016/j.htct.2022.11.006. Retrieved November 2025.
- Liu, B., Zhou, H., Tan, L., Siu, K. T. H., & Guan, X.-Y. (2024, July 17). Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduction and Targeted Therapy, 9(1), 175. https://doi.org/10.1038/s41392-024-01856-7. Retrieved November 2025.
- Sarivalasis, A., Boudousquié, C., Balint, K., Stevenson, B. J., Gannon, P. O., Iancu, E. M., … Kandalaft, L. E. (2019, November 26). A Phase I/II trial comparing autologous dendritic cell vaccine pulsed either with personalized peptides (PEP-DC) or with tumor lysate (OC-DC) in patients with advanced high-grade ovarian serous carcinoma. Journal of Translational Medicine, 17(1), 391. https://doi.org/10.1186/s12967-019-02133-w. Retrieved November 2025.
- Wang, Y., et al. (2020). Personalized neoantigen-pulsed dendritic cell vaccine for advanced lung cancer. Signal Transduction and Targeted Therapy, 5, 57. https://doi.org/10.1038/s41392-020-00448-5. Retrieved November 2025.
- Al Saihati, H. A. (2021, December). Overview of Dendritic Cell Vaccines as Effective Approaches in Cancer Immunotherapy. Bahrain Medical Bulletin, 43(4), 737–741. Retrieved November 2025.
- van de Loosdrecht, A. A., van Wetering, S., Santegoets, S. J. A. M., Singh, S. K., Eeltink, C. M., den Hartog, Y., … de Gruijl, T. D. (2019). A novel allogeneic off-the-shelf dendritic cell vaccine for post-remission treatment of elderly patients with acute myeloid leukemia. Cancer Immunology, Immunotherapy. https://doi.org/10.1007/s00262-018-2198-9. Retrieved November 2025.
- Plumas, J. (2022). Harnessing dendritic cells for innovative therapeutic cancer vaccines. Current Opinion in Oncology, 34, 161–168. https://doi.org/10.1097/CCO.0000000000000815. Retrieved November 2025.
- Moseman, J. E., Shim, D., Jeon, D., Rastogi, I., Schneider, K. M., & McNeel, D. G. (2025, September 14). Messenger RNA and Plasmid DNA Vaccines for the Treatment of Cancer. Vaccines, 13(9), 976. https://doi.org/10.3390/vaccines13090976. Retrieved November 2025.
- Sehgal, K., Dhodapkar, K. M., & Dhodapkar, M. V. (2014). Targeting human dendritic cells in situ to improve vaccines. Immunology Letters. https://doi.org/10.1016/j.imlet.2014.05.005. Retrieved November 2025.
- Zhang, S. (2025). Dendritic cell vaccines: Current research progress. ScienceDirect (journal details via ScienceDirect article page). Retrieved November 2025.
- ScienceDirect. (2025). Dendritic Cell Function – an overview. ScienceDirect Topics. Retrieved November 2025.
- European Medicines Agency (EMA). (2025, March 13). Unregulated advanced therapy medicinal products pose serious risks to health. Retrieved October 2025.
- Barrie, R. (2025, March 14). Unsafe and unregulated cell therapies being sold in Europe. Pharmaceutical Technology. Retrieved September 2025.
- Gupta, S., Carballido, E., & Fishman, M. (2011). Sipuleucel-T for therapy of asymptomatic or minimally symptomatic, castrate-refractory prostate cancer: an update and perspective among other treatments. OncoTargets and Therapy, 4, 79–96. https://doi.org/10.2147/OTT.S14107. Retrieved November 2025.
- Mastelic-Gavillet, B., Balint, K., Boudousquie, C., Gannon, P. O., & Kandalaft, L. E. (2019, April 11). Personalized Dendritic Cell Vaccines—Recent Breakthroughs and Encouraging Clinical Results. Frontiers in Immunology, 10, 766. https://doi.org/10.3389/fimmu.2019.00766. Retrieved November 2025.
- AdisInsight, Springer Nature. (2021). Autologous dendritic cell immunotherapy – APAC Biotech. AdisInsight Drug Profile (APCEDEN). Latest information update 31 October 2021. Retrieved October 2025.
- Director for Approval Management. (2021, April). 2020 Drug Approval Report. Retrieved November 2025.
- Dejaegher, J., Van Gool, S., & De Vleeschouwer, S. (2014, March 13). Dendritic cell vaccination for glioblastoma multiforme: Review with focus on predictive factors for treatment response. ImmunoTargets and Therapy, 3, 55–66. https://doi.org/10.2147/ITT.S40121. Retrieved September 2025.
- Liau, L. M., Ashkan, K., Tran, D. D., Campian, J. L., Trusheim, J. E., Cobbs, C. S., … Bosch, M. L. (2018, May 29). First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. Journal of Translational Medicine, 16, 142. https://doi.org/10.1186/s12967-018-1507-6. Retrieved November 2025.
- Vreeland, T. J., Clifton, G. T., Hale, D. F., Chick, R. C., Hickerson, A. T., Cindass, J. L., … Faries, M. B. (2021, February 27). A Phase IIb randomized controlled trial of the TLPLDC vaccine as adjuvant therapy after surgical resection of stage III/IV melanoma: A primary analysis. Annals of Surgical Oncology, 28(11), 6126–6137. https://doi.org/10.1245/s10434-021-09709-1. Retrieved November 2025.
- van Willigen, W. W., Bloemendal, M., Boers-Sonderen, M. J., de Groot, J. W. B., Koornstra, R. H. T., van der Veldt, A. A. M., … Bol, K. F. (2020, March 22). Response and survival of metastatic melanoma patients treated with immune checkpoint inhibition for recurrent disease on adjuvant dendritic cell vaccination. OncoImmunology, 9(1), 1738814. https://doi.org/10.1080/2162402X.2020.1738814. Retrieved November 2025.
- Sprooten, J., Ceusters, J., Coosemans, A., Agostinis, P., De Vleeschouwer, S., Zitvogel, L., … Garg, A. D. (2019, July 18). Trial watch: Dendritic cell vaccination for cancer immunotherapy. OncoImmunology, 8(11), e1638212. https://doi.org/10.1080/2162402X.2019.1638212. Retrieved November 2025.
- Hanul, A. V., Khranovska, N. M., Sovenko, V. M., Hanul, V. L., Hrynevych, Yu. A., Orel, V. E., … Kondratskyi, Yu. M. (2012). Experience of using dendritic cell autovaccine in treatment of patients with non-small cell lung cancer. Clinical Oncology, 7(3), 1–4. Retrieved November 2025.
- Qian, D., Li, J., Huang, M., Cui, Q., Liu, X., & Sun, K. (2023, June). Dendritic cell vaccines in breast cancer: Immune modulation and immunotherapy. Biomedicine & Pharmacotherapy, 162, 114685. https://doi.org/10.1016/j.biopha.2023.114685. Retrieved September 2025.
- Zhang, X., He, T., Li, Y., Chen, L., Liu, H., Wu, Y., & Guo, H. (2021, January 25). Dendritic cell vaccines in ovarian cancer. Frontiers in Immunology, 11, 613773. https://doi.org/10.3389/fimmu.2020.613773. Retrieved October 2025.
- Berntsen, A., Trepiakas, R., Wenandy, L., Geertsen, P. F., thor Straten, P., Andersen, M. H., … Svane, I. M. (2008, October). Therapeutic dendritic cell vaccination of patients with metastatic renal cell carcinoma: A clinical phase 1/2 trial. Journal of Immunotherapy, 31(8), 771–780. https://doi.org/10.1097/CJI.0b013e3181833818. Retrieved November 2025.
- Wang, B., Wu, K., Cui, Y., Han, X., & Xing, T. (2025, September). Effects of a dendritic cell vaccine loaded with whole tumor antigen on bladder cancer model in hu-PBL-SCID mice. Drug Development Research, 86(6), e70157. https://doi.org/10.1002/ddr.70157. Retrieved September 2025.
- The ASCO Post Staff. (2025, August 28). Adding Dendritic Cell Vaccination to Hepatocellular Carcinoma Therapy. The ASCO Post. Retrieved November 2025.
- Dafni, U., Martin-Lluesma, S., Balint, K., Tsourti, Z., Vervita, K., Chenal, J., … Kandalaft, L. E. (2021, January). Efficacy of cancer vaccines in selected gynaecological breast and ovarian cancers: A 20-year systematic review and meta-analysis. European Journal of Cancer, 142, 63–82. https://doi.org/10.1016/j.ejca.2020.10.014. Retrieved November 2025.
- Iamukova, L., & Alferova, E. (2025, August 13). Personalized cancer vaccines in the clinical trial pipeline. Asia-Pacific Journal of Clinical Oncology. Advance online publication. https://doi.org/10.1111/ajco.70006. Retrieved November 2025.
- Anguille, S., Van de Velde, A. L., Smits, E. L., Van Tendeloo, V. F., Cools, N., Nijs, G., … Van Driessche, A. (2017, October 12). Dendritic cell vaccination as postremission treatment to prevent or delay relapse in acute myeloid leukemia. Blood, 130(15), 1713–1721. https://doi.org/10.1182/blood-2017-04-780155. Retrieved November 2025.
- Westermann, J., Kopp, J., Körner, I., Kurz, S., Zenke, M., … Dörken, B. (2000, May). Bcr/abl+ autologous dendritic cells for vaccination in chronic myeloid leukemia. Bone Marrow Transplantation, 25(Suppl 2), S46–S49. https://doi.org/10.1038/sj.bmt.1702354. Retrieved November 2025.
- Palma, M., Adamson, L., Hansson, L., Kokhaei, P., Rezvany, R., Mellstedt, H., … Choudhury, A. (2008, November). Development of a dendritic cell-based vaccine for chronic lymphocytic leukemia. Cancer Immunology, Immunotherapy, 57, 1705–1710. https://doi.org/10.1007/s00262-008-0561-y. Retrieved November 2025.
- Weinstock, M., Rosenblatt, J., & Avigan, D. (2017). Dendritic cell therapies for hematologic malignancies. Molecular Therapy – Methods & Clinical Development, 5, 66–75. https://doi.org/10.1016/j.omtm.2017.03.004. Retrieved November 2025.
- Reichardt, V. L., & Brossart, P. (2005). Dendritic cells in clinical trials for multiple myeloma. Methods in Molecular Medicine, 109, 127–136. https://doi.org/10.1385/1-59259-862-5:127. Retrieved November 2025.
- Büchler, T., Michalek, J., Kovarova, L., Musilova, R., & Hajek, R. (2003, April). Dendritic cell-based immunotherapy for the treatment of hematological malignancies. Hematology, 8(2), 97–104. https://doi.org/10.1080/1024533031000084204. Retrieved November 2025.
- Eyrich, M., Rachor, J., Schreiber, S. C., Wölfl, M., & Schlegel, P. G. (2013, June 10). Dendritic cell vaccination in pediatric gliomas: Lessons learnt and future perspectives. Frontiers in Pediatrics, 1, 12. https://doi.org/10.3389/fped.2013.00012. Retrieved November 2025.
- National Library of Medicine (U.S.). (2012). Dendritic Cell Vaccine for Children and Adults With Sarcoma (NCT01803152). ClinicalTrials.gov. Retrieved November 2025.
- Schnitger, A. (2014). The Hospital Exemption, a regulatory option for unauthorised advanced therapy medicinal products. Master’s thesis, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany. Retrieved November 2025.
- Draube, A., Klein-González, N., Mattheus, S., Brillant, C., Ziske, C., et al. (2011, April 20). Dendritic cell based tumor vaccination in prostate and renal cell cancer: A systematic review and meta-analysis. PLOS ONE, 6(4), e18801. https://doi.org/10.1371/journal.pone.0018801. Retrieved November 2025.
- Gerritsen, W. R., & Sharma, P. (2012). Current and emerging treatment options for castration-resistant prostate cancer: A focus on immunotherapy. Journal of Clinical Immunology, 32(1), 25–35. https://doi.org/10.1007/s10875-011-9595-6. Retrieved November 2025.
- Kawalec, P., Paszulewicz, A., Holko, P., & Pilc, A. (2012, October 31). Sipuleucel-T immunotherapy for castration-resistant prostate cancer: A systematic review and meta-analysis. Archives of Medical Science, 8(5), 767–775. https://doi.org/10.5114/aoms.2012.31610. Retrieved November 2025.
- Polyzoidis, S., & Ashkan, K. (2014). DCVax-L—developed by Northwest Biotherapeutics. Human Vaccines & Immunotherapeutics, 10(11), 3139–3145. https://doi.org/10.4161/hv.29276. Retrieved November 2025.
- Rahma, O. E., Gammoh, E., Simon, R. M., & Khleif, S. N. (2014, September). Is the “3+3” dose-escalation phase I clinical trial design suitable for therapeutic cancer vaccine development? A recommendation for alternative design. Clinical Cancer Research, 20(18), 4758–4767. https://doi.org/10.1158/1078-0432.CCR-13-2671. Retrieved November 2025.
- British Society for Immunology. Dendritic Cells: Migration. BiteSized Immunology – Systems & Processes. Retrieved November 2025.
- Bapsy, P. P., Sharan, B., Kumar, C., Das, R. P., Rangarajan, B., Jain, M., … Srivastava, M. (2014). Open-label, multi-center, non-randomized, single-arm study to evaluate the safety and efficacy of dendritic cell immunotherapy in patients with refractory solid malignancies, on supportive care. Cytotherapy, 16, 234–244. https://doi.org/10.1016/j.jcyt.2013.11.013. Retrieved November 2025.
- Boudewijns, S., Bloemendal, M., Gerritsen, W. R., de Vries, I. J. M., & Schreibelt, G. (2016, October 2). Dendritic cell vaccination in melanoma patients: From promising results to future perspectives. Human Vaccines & Immunotherapeutics, 12(10), 2523–2528. https://doi.org/10.1080/21645515.2016.1197453. Retrieved November 2025.
- Hickerson, A., Clifton, G. T., Brown, T. A., Campf, J., Myers, J. W., Vreeland, T. J., … Faries, M. B. (2019, May). Clinical efficacy of vaccination with the autologous tumor lysate particle loaded dendritic cell (TLPLDC) vaccine in metastatic melanoma. Journal of Clinical Oncology, 37(15_suppl), e21025. https://doi.org/10.1200/JCO.2019.37.15_suppl.e21025. Retrieved November 2025.
- Zhang, L., Yang, X., Sun, Z., Li, J., Zhu, H., Li, J., Pang, Y. (2016, February 24). Dendritic cell vaccine and cytokine-induced killer cell therapy for the treatment of advanced non-small cell lung cancer. Oncology Letters, 11(4), 2605–2610. https://doi.org/10.3892/ol.2016.4273. Retrieved November 2025.
- Stevens, D., Ingels, J., Van Lint, S., Vandekerckhove, B., Vermaelen, K. (2021, February 12). Dendritic Cell-Based Immunotherapy in Lung Cancer. Frontiers in Immunology, 11, 620374. https://doi.org/10.3389/fimmu.2020.620374. Retrieved November 2025.
- Kobayashi, M., Chiba, A., Izawa, H., Yanagida, E., Okamoto, M., Shimodaira, S., Yonemitsu, Y., Shibamoto, Y., Suzuki, N., Nagaya, M. (2014, May 7). The feasibility and clinical effects of dendritic cell-based immunotherapy targeting synthesized peptides for recurrent ovarian cancer. Journal of Ovarian Research, 7(1), 48. https://doi.org/10.1186/1757-2215-7-48. Retrieved November 2025.
- Jiang, J., Li, L., Yu, Z., Wang, K., Ma, J., Li, D., Guo, Z., Wang, L., Zhang, H., Xu, L., Zhang, M. (2025, October 20). Clinical efficacy and challenges of cancer-testis antigen vaccines in advanced solid tumors: A systematic review and meta-analysis. Critical Reviews in Oncology/Hematology, 216, 104981. https://doi.org/10.1016/j.critrevonc.2025.104981. Retrieved November 2025.
- Han, C.-L., Yan, Y.-C., Yan, L.-J., Meng, G.-X., Yang, C.-C., Liu, H., … Li, T. (2022, April 28). Efficacy and security of tumor vaccines for hepatocellular carcinoma: A systemic review and meta-analysis of the last 2 decades. Journal of Cancer Research and Clinical Oncology, 149(4), 1425–1441. https://doi.org/10.1007/s00432-022-04008-y. Retrieved November 2025.
- Wainberg, Z. A., Weekes, C. D., Furqan, M., Devoe, C. E., Leal, A. D., Chung, V., … O’Reilly, E. M. (2025, November). Lymph node-targeted, mKRAS-specific amphiphile vaccine in pancreatic and colorectal cancer: Phase 1 AMPLIFY-201 trial final results. Nature Medicine, 31(11), 3648–3653. https://doi.org/10.1038/s41591-025-03876-4. Retrieved November 2025.
- Schuler, P. J., Harasymczuk, M., Visus, C., De Leo, A., Trivedi, S., Lei, Y., et al. (2014, February 28). Phase I dendritic cell p53 peptide vaccine for head and neck cancer. Clinical Cancer Research, 20(9), 2433–2444. https://doi.org/10.1158/1078-0432.CCR-13-2617. Retrieved November 2025.
- Eckl, J., Raffegerst, S., Schnorfeil, F., et al. (2019, December 9). DC vaccination induces antigen-specific immune responses in AML patients: A 1-year interim assessment. Blood, 134(Supplement 1), 3923. https://doi.org/10.1182/blood-2019-129749. Retrieved November 2025.
- Rosenblatt, J., Vasir, B., Uhl, L., Blotta, S., Macnamara, C., Somaiya, P., … Avigan, D. (2011, January 13). Vaccination with dendritic cell/tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma. Blood, 117(2), 393–402. https://doi.org/10.1182/blood-2010-04-277137. Retrieved November 2025.
- Caruso, D. A., Orme, L. M., Neale, A. M., Radcliff, F. J., Amor, G. M., Maixner, W., … Ashley, D. M. (2004, July). Results of a phase 1 study utilizing monocyte-derived dendritic cells pulsed with tumor RNA in children and young adults with brain cancer. Neuro-Oncology, 6(3), 236–246. https://doi.org/10.1215/S1152851703000668. Retrieved November 2025.
- Geiger, J. D., Hutchinson, R. J., Hohenkirk, L. F., McKenna, E. A., Yanik, G. A., Levine, J. E., Chang, A. E., Braun, T. M., & Mulé, J. J. (2001, December 1). Vaccination of pediatric solid tumor patients with tumor lysate-pulsed dendritic cells can expand specific T cells and mediate tumor regression. Cancer Research, 61(23), 8513–8519. Retrieved November 2025.
- Dillman, R. O., Cornforth, A. N., Nistor, G. I., McClay, E. F., Amatruda, T. T., & Depriest, C. (2018, March 6). Randomized phase II trial of autologous dendritic cell vaccines versus autologous tumor cell vaccines in metastatic melanoma: 5-year follow up and additional analyses. Journal for ImmunoTherapy of Cancer, 6(1), 19. https://doi.org/10.1186/s40425-018-0330-1. Retrieved November 2025.
- Kantoff, P. W., Higano, C. S., Shore, N. D., Berger, E. R., Small, E. J., Penson, D. F., Redfern, C. H., Ferrari, A. C., Dreicer, R., Sims, R. B., Xu, Y., Frohlich, M. W., & Schellhammer, P. F. (2010, July 29). Sipuleucel-T immunotherapy for castration-resistant prostate cancer. New England Journal of Medicine, 363(5), 411–422. https://doi.org/10.1056/NEJMoa1001294. Retrieved November 2025.
- Madan, R. A., Antonarakis, E. S., Drake, C. G., Fong, L., Yu, E. Y., McNeel, D. G., Lin, D. W., Chang, N. N., Sheikh, N. A., & Gulley, J. L. (2020, June 1). Putting the pieces together: Completing the mechanism of action jigsaw for sipuleucel-T. Journal of the National Cancer Institute, 112(6), 562–573. https://doi.org/10.1093/jnci/djaa021. Retrieved November 2025.
- Boudewijns, S. (2017). Dendritic cell vaccination in the evolving therapeutic landscape of melanoma. Doctoral dissertation, Radboud University Medical Center, Nijmegen, The Netherlands. Retrieved November 2025.
- Medscape. Non-Small Cell Lung Cancer (NSCLC): Treatment & management. Retrieved November 2025.
- American Cancer Society (ACS). (2022, April 12). Treatment of Breast Cancer Stages I–III. Retrieved November 2025.
- Inogés, S., Tejada, S., de Cerio, A. L. D., Pastor, F., Villanueva, H., Gallego Pérez-Larraya, J., … Bendandi, M. (2017, May). A phase II trial of autologous dendritic cell vaccination and radiochemotherapy following fluorescence-guided surgery in newly diagnosed glioblastoma patients. Journal of Translational Medicine, 15, 104. https://doi.org/10.1186/s12967-017-1202-z. Retrieved November 2025.
- Rutkowski, S., De Vleeschouwer, S., Kaempgen, E., Wolff, J. E. A., Kühl, J., Demaerel, P., … Van Gool, S. W. (2004, November 2). Surgery and adjuvant dendritic cell-based tumour vaccination for patients with relapsed malignant glioma, a feasibility study. British Journal of Cancer, 91(9), 1656–1662. https://doi.org/10.1038/sj.bjc.6602195. Retrieved November 2025.
- American Cancer Society (ACS). (2025, July 21). Living with Advanced and Metastatic Cancer: Support and Symptom Management. Retrieved November 2025.
- Mollica, M. A., Tesauro, G. M., Mao, J. J., McCabe, M. S., Tonorezos, E. S., & Jacobsen, P. B. (2021, May 1). Survivorship for individuals living with advanced and metastatic cancer: National Cancer Institute meeting report. American Journal of Respiratory and Critical Care Medicine, 203(9), 1103–1110. https://doi.org/10.1164/rccm.201508-1573OC. Retrieved November 2025.
- Schneble, E. J., Yu, X., Wagner, T. E., & Peoples, G. E. (2014). Novel dendritic cell-based vaccination in late stage melanoma. Human Vaccines & Immunotherapeutics, 10(11), 3132–3138. https://doi.org/10.4161/hv.29110. Retrieved November 2025.
- Piccioni, D. E., Duma, C. M., Kesari, S., LaRocca, R. V., Aiken, R. D., Taylor, T. H., Carrillo, J. A., Abedi, M., Nistor, G. I., Keirstead, H. S., Dillman, R. O., & Bota, D. A. (2022, September 30). Enhanced progression-free survival in a phase 2 trial of personal dendritic cell vaccines in patients with newly diagnosed glioblastoma. Journal of Oncology Research and Therapy, 7, 10149. https://doi.org/10.29011/2574-710X.010149. Retrieved November 2025.
- Bota, D. A., Taylor, T. H., Piccioni, D. E., Duma, C. M., LaRocca, R. V., Kesari, S., Carrillo, J. A., Abedi, M., Aiken, R. D., Hsu, F. P. K., Kong, X.-T., Hsieh, C., Bota, P. G., Nistor, G. I., Keirstead, H. S., & Dillman, R. O. (2022, December 14). Phase 2 study of AV-GBM-1 (a tumor-initiating cell targeted dendritic cell vaccine) in newly diagnosed Glioblastoma patients: safety and efficacy assessment. Journal of Experimental & Clinical Cancer Research, 41, 344. https://doi.org/10.1186/s13046-022-02552-6. Retrieved November 2025.
- Liau, L. M., Ashkan, K., Brem, S., et al. (2022, November 17). Association of autologous tumor lysate-loaded dendritic cell vaccination with extension of survival among patients with newly diagnosed and recurrent glioblastoma: A phase 3 prospective externally controlled cohort trial. JAMA Oncology, 9(1), 112–121. https://doi.org/10.1001/jamaoncol.2022.5370. Retrieved November 2025.
- National Library of Medicine (U.S.). Study of a drug [DCVax-L] to treat newly diagnosed GBM brain cancer (NCT00045968). ClinicalTrials.gov. Retrieved November 2025.
- Datsi, A., & Sorg, R. V. (2021, November 2). Dendritic Cell Vaccination of Glioblastoma: Road to Success or Dead End. Frontiers in Immunology, 12, 770390. https://doi.org/10.3389/fimmu.2021.770390. Retrieved November 2025.
- Van Gool, S. W., Makalowski, J., Kampers, L. F. C., Van de Vliet, P., Sprenger, T., Schirrmacher, V., & Stücker, W. (2023). Dendritic cell vaccination for glioblastoma multiforme patients: has a new milestone been reached?. Translational Cancer Research, 12(8), 2224–2228. https://doi.org/10.21037/tcr-23-603. Retrieved November 2025.
- Akutsu, Y., Matsubara, H., Urashima, T., Komatsu, A., Sakata, H., Nishimori, T., Yoneyama, Y., Hoshino, I., Murakami, K., Usui, A., Kano, M., & Ochiai, T. (2007, September). Combination of direct intratumoral administration of dendritic cells and irradiation induces strong systemic antitumor effect mediated by GRP94/gp96 against squamous cell carcinoma in mice. International Journal of Oncology, 31(3), 509–515. https://doi.org/10.3892/ijo.31.3.509. Retrieved November 2025.
- Formenti, S. C., & Demaria, S. (2012, November 15). Radiation therapy to convert the tumor into an in situ vaccine. International Journal of Radiation Oncology, Biology, Physics, 84(4), 879–880. https://doi.org/10.1016/j.ijrobp.2012.06.020. Retrieved November 2025.
- World Health Organization (WHO). (2021, March 5). New WHO/IAEA publication provides guidance on radiotherapy equipment to fight cancer. Retrieved November 2025.
- Jiang, W., Wang, Z., Luo, Q., Dai, Z., Zhu, J., Tao, X., et al. (2024, December 20). Combined immunotherapy with dendritic cells and cytokine-induced killer cells for solid tumors: A systematic review and meta-analysis of randomized controlled trials. Journal of Translational Medicine, 22, 1122. https://doi.org/10.1186/s12967-024-05940-y. Retrieved November 2025.
- Gao, D., Li, C., Xie, X., Zhao, P., Wei, X., Sun, W., et al. (2014, April 3). Autologous tumor lysate-pulsed dendritic cell immunotherapy with cytokine-induced killer cells improves survival in gastric and colorectal cancer patients. PLOS ONE, 9(4), e93886. https://doi.org/10.1371/journal.pone.0093886. Retrieved November 2025.
- Zhou, X., Mo, X., Qiu, J., Zhao, J., Wang, S., Zhou, C., et al. (2018, November 5). Chemotherapy combined with dendritic cell vaccine and cytokine-induced killer cells in the treatment of colorectal carcinoma: A meta-analysis. Cancer Management and Research, 10, 5363–5372. https://doi.org/10.2147/CMAR.S173201. Retrieved November 2025.
- Yu, J., Sun, H., Cao, W., Song, Y., Jiang, Z., et al. (2022, January 24). Research progress on dendritic cell vaccines in cancer immunotherapy. Experimental Hematology & Oncology, 11, 3. https://doi.org/10.1186/s40164-022-00257-2. Retrieved November 2025.
- Vansteenkiste, J., Demedts, I., Cuppens, K., Pons-Tostivint, E., Wauters, E., Borm, F. J., et al. (2025, October). Innovative therapeutic cancer vaccine PDC*lung01 with or without anti-PD-1: An open-label, dose-escalation phase I/II study in non-small-cell lung cancer. ESMO Open, 10(11), 105844. https://doi.org/10.1016/j.esmoop.2025.105844. Retrieved November 2025.
- Jeng, L.-B., Liao, L.-Y., Shih, F.-Y., & Teng, C.-F. (2022, September 8). Dendritic-Cell-Vaccine-Based Immunotherapy for Hepatocellular Carcinoma: Clinical Trials and Recent Preclinical Studies. Cancers, 14(18), 4380. https://doi.org/10.3390/cancers14184380. Retrieved November 2025.
- Rizell, M., Sternby Eilard, M., Andersson, M., Andersson, B., Karlsson-Parra, A., & Suenaert, P. (2019, January 21). Phase 1 Trial With the Cell-Based Immune Primer Ilixadencel, Alone, and Combined With Sorafenib, in Advanced Hepatocellular Carcinoma. Frontiers in Oncology, 9, 19. https://doi.org/10.3389/fonc.2019.00019. Retrieved November 2025.
- Wang, L., Li, X., Dong, X.-J., Yu, X.-L., Zhang, J., Cheng, Z.-G., … Liang, P. (2024). Dendritic cell-cytokine killer combined with microwave ablation reduced recurrence for hepatocellular carcinoma compared to ablation alone. Technology and Health Care, 32(3), 1819–1834. https://doi.org/10.3233/THC-230871. Retrieved November 2025.
- Buonaguro, L., Mauriello, A., Cavalluzzo, B., Petrizzo, A., & Tagliamonte, M. (2019, March–April). Immunotherapy in hepatocellular carcinoma. Annals of Hepatology, 18(2), 291–297. https://doi.org/10.1016/j.aohep.2019.04.003. Retrieved November 2025.
- U.S. Food and Drug Administration (FDA). (2019, May 28). PROVENGE (sipuleucel-T) prescribing information. Retrieved November 2025.
- Lichtenegger, F. S., Krupka, C., Haubner, S., Köhnke, T., & Subklewe, M. (2017, July 25). Recent developments in immunotherapy of acute myeloid leukemia. Journal of Hematology & Oncology, 10(1), 142. https://doi.org/10.1186/s13045-017-0505-0. Retrieved November 2025.
- Van Tendeloo, V. F., Van de Velde, A., Van Driessche, A., Cools, N., Anguille, S., Ladell, K., … Berneman, Z. N. (2010, August 3). Induction of complete and molecular remissions in acute myeloid leukemia by Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proceedings of the National Academy of Sciences of the United States of America, 107(31), 13824–13829. https://doi.org/10.1073/pnas.1008051107. Retrieved November 2025.
- Castiello, L., Aricò, E., D’Agostino, G., Santodonato, L., & Belardelli, F. (2019, September 25). In situ vaccination by direct dendritic cell inoculation: The coming of age of an old idea?. Frontiers in Immunology, 10, 2303. https://doi.org/10.3389/fimmu.2019.02303. Retrieved November 2025.
- Ingels, J., De Cock, L., Stevens, D., Mayer, R. L., Théry, F., Sanchez Sanchez, G., … Vandekerckhove, B. (2024, May 21). Neoantigen-targeted dendritic cell vaccination in lung cancer patients induces long-lived T cells exhibiting the full differentiation spectrum. Cell Reports Medicine, 5(5), 101516. https://doi.org/10.1016/j.xcrm.2024.101516. Retrieved November 2025.
- UiO Faculty of Medicine, CanCell – Centre for Cancer Cell Reprogramming. (2023, August 23). Dendritic cell vaccine as maintenance therapy for AML. Retrieved November 2025.
- Subklewe, M., Geiger, C., Lichtenegger, F. S., Javorovic, M., Kvalheim, G., Schendel, D. J., & Bigalke, I. (2014, October). New generation dendritic cell vaccine for immunotherapy of acute myeloid leukemia. Cancer Immunology, Immunotherapy, 63(10), 1093–1103. https://doi.org/10.1007/s00262-014-1600-5. Retrieved November 2025.
- Anguille, S., Smits, E. L., Lion, E., Van Tendeloo, V. F., & Berneman, Z. N. (2014, June). Clinical use of dendritic cells for cancer therapy. The Lancet Oncology, 15(7), e257–e267. https://doi.org/10.1016/S1470-2045(13)70585-0. Retrieved November 2025.
- Van Willigen, W. W., Bloemendal, M., Gerritsen, W. R., Schreibelt, G., & de Vries, I. J. M. (2018, October 1). Dendritic Cell Cancer Therapy: Vaccinating the Right Patient at the Right Time. Frontiers in Immunology, 9, 2265. https://doi.org/10.3389/fimmu.2018.02265. Retrieved November 2025.
- Maeng, H. M., Moore, B. N., Bagheri, H., Steinberg, S. M., Inglefield, J., Dunham, K., … Berzofsky, J. A. (2021, December 16). Phase I Clinical Trial of an Autologous Dendritic Cell Vaccine Against HER2 Shows Safety and Preliminary Clinical Efficacy. Frontiers in Oncology, 11, 789078. https://doi.org/10.3389/fonc.2021.789078. Retrieved November 2025.
- Ridolfi, L., Gurrieri, L., Riva, N., Bulgarelli, J., De Rosa, F., Guidoboni, M., … Tosatto, L. (2024, August 13). First step results from a phase II study of a dendritic cell vaccine in glioblastoma patients (CombiG-vax). Frontiers in Immunology, 15, 1404861. https://doi.org/10.3389/fimmu.2024.1404861. Retrieved November 2025.
- Cancer Research UK. (2024, April 23). Treatment to remove white blood cells (leukapheresis). Retrieved November 2025.
- St. Jude Children’s Research Hospital. Leukapheresis – Together by St. Jude. Retrieved November 2025.
- Encyclopaedia Britannica. Leukapheresis. Retrieved November 2025.
- Yi, Q., & Kwak, L. W. (2005, February 1). Monocyte-derived dendritic cells: A promising armament for immunotherapy in human malignancies. Clinical Cancer Research, 11(3), 966–967. https://doi.org/10.1158/1078-0432.CCR-04-1911. Retrieved November 2025.
- Van Acker, H. H., Anguille, S., Willemen, Y., Smits, E. L. J., Van Tendeloo, V. F. I., & Berneman, Z. N. (2020, January 17). Development and optimization of a GMP-compliant manufacturing process for a personalized tumor lysate dendritic cell vaccine. Vaccines, 8(1), 25. https://doi.org/10.3390/vaccines8010025. Retrieved November 2025.
- Papakonstantinou, A., Marinos, G., Giannakoulas, N., Papadopoulos, N., Douroudis, K., & Tsoulfas, G. (2025). The efficacy of neoantigen-loaded dendritic cell vaccine immunotherapy in non-metastatic gastric cancer. Medicina, 13(3), 90. https://doi.org/10.3390/medicina13030090. Retrieved November 2025.
- Mishchenko, T. A., Sleptsova, E. E., Turubanova, V. D., Savyuk, M. O., Lermontova, S. A., Klapshina, L. G., … Krysko, D. V. (2023, October). Dendritic cells pulsed with tumor lysates induced by tetracyanotetra (aryl) porphyrazines-based photodynamic therapy effectively trigger anti-tumor immunity in an orthotopic mouse glioma model. Pharmaceutics, 15(10), 2430. https://doi.org/10.3390/pharmaceutics15102430. Retrieved November 2025.
- Dillman, R. O., & Depriest, C. (2018, September 26). Dendritic cell vaccines presenting autologous tumor antigens from self-renewing cancer cells in metastatic renal cell carcinoma. Journal of Exploratory Research in Pharmacology, 3(4), 96–104. https://doi.org/10.14218/JERP.2018.00012. Retrieved November 2025.
- Reinhard, G., Märten, A., Kiske, S. M., Feil, F., Bieber, T., & Schmidt-Wolf, I. G. H. (2002, May 20). Generation of dendritic cell-based vaccines for cancer therapy. British Journal of Cancer, 86(10), 1529–1533. https://doi.org/10.1038/sj.bjc.6600316. Retrieved November 2025.
- Leal, L., Couto, E., Sánchez-Palomino, S., Climent, N., Fernández, I., Miralles, L., … García, F. (2021, November 11). Effect of intranodally administered dendritic cell-based HIV vaccine in combination with pegylated interferon α-2a on viral control following ART discontinuation: A phase 2A randomized clinical trial. Frontiers in Immunology, 12, 767370. https://doi.org/10.3389/fimmu.2021.767370. Retrieved November 2025.