A new approach to overcoming treatment resistance in relapsed/refractory AML and B-cell malignancies
Imagine being told that the life-saving cancer treatment you received has stopped working, and your disease has returned. For patients with relapsed or refractory acute myeloid leukemia (AML) and B-cell malignancies, this devastating scenario is all too common. The problem often lies in a clever survival protein called Mcl-1 that cancer cells use to resist treatment. When targeted therapies like venetoclax successfully block other survival pathways, cancer cells increasingly rely on Mcl-1 to stay alive 1 4 . This resistance mechanism has represented a significant challenge in hematologyâuntil now.
A key survival protein that cancer cells use to resist treatment and evade cell death.
A novel approach to indirectly target Mcl-1 by disrupting its production mechanism.
Recent research brings new hope through an innovative approach: indirectly targeting Mcl-1 by inhibiting cyclin-dependent kinase 9 (CDK9), a key regulator of protein production in cells. In a landmark phase 1 clinical trial published in Blood Advances, researchers have demonstrated that an experimental oral drug called voruciclib can safely disrupt this resistance pathway in patients who have exhausted other treatment options 1 3 8 . This breakthrough represents a potentially promising strategy in the ongoing battle against treatment-resistant blood cancers.
To understand voruciclib's potential, we first need to examine Mcl-1 (myeloid cell leukemia-1), a protein that plays a crucial role in cancer cell survival. Mcl-1 belongs to the Bcl-2 family of proteins that regulate programmed cell death (apoptosis). Think of these proteins as switches that control when a cell should self-destructâa natural process that prevents damaged cells from multiplying.
In healthy cells, this system maintains balance. But in cancers like AML and certain B-cell malignancies, cancer cells produce excessive Mcl-1, effectively jamming the "off" switch on their self-destruct mechanism. This allows them to survive indefinitely and resist chemotherapy 4 7 . Even when targeted drugs like venetoclax successfully block other survival proteins (specifically Bcl-2), cancer cells can compensate by increasing their reliance on Mcl-1, leading to treatment resistance 1 4 .
This is where CDK9 inhibition comes in. Rather than directly targeting the Mcl-1 protein itselfâan approach that has proven challenging due to safety concernsâresearchers have taken an indirect route 4 .
CDK9 acts as a master regulator of transcription, controlling the production of short-lived proteins, including Mcl-1. By inhibiting CDK9, voruciclib essentially reduces the factory output of Mcl-1 and other cancer-promoting proteins, effectively pulling the rug out from under the cancer cell's resistance strategy 5 .
This approach offers a strategic advantage: by targeting the production mechanism rather than the protein itself, voruciclib can simultaneously reduce multiple cancer-promoting proteins that share the characteristic of being short-lived, including MYC and NF-κB 4 8 .
CDK9 forms a complex with cyclin T to create P-TEFb, which phosphorylates RNA polymerase II to initiate transcription of short-lived proteins like Mcl-1.
Voruciclib binds to CDK9, inhibiting its kinase activity and preventing phosphorylation of RNA polymerase II.
With transcription disrupted, cancer cells cannot produce sufficient Mcl-1 to maintain their survival advantage.
Depleted Mcl-1 levels allow the natural cell death process to resume, leading to cancer cell elimination.
The phase 1 dose-escalation study, registered as #NCT03547115 at ClinicalTrials.gov, enrolled 40 patients with relapsed or refractory blood cancersâ21 with AML and 19 with various B-cell malignancies 1 4 . These were heavily pretreated patients, having received a median of 3 prior lines of therapy (range: 1-8), representing a population with limited treatment options 1 8 .
The study followed a dose-escalation design to determine the safety, tolerability, and optimal dosing schedule of voruciclib. The initial plan was to administer voruciclib daily on a continuous 28-day cycle (Group 1). However, the researchers demonstrated adaptive design principles when two patients who had previously undergone stem cell transplantation developed interstitial pneumonitis (lung inflammation) at the 100 mg dose level 1 4 8 .
This observation led to a protocol adjustment: subsequent patients (Group 2) received voruciclib on an intermittent schedule (days 1-14 of a 28-day cycle), which significantly improved the safety profile 4 7 .
Patients Enrolled
The primary goal of this phase 1 trial was to identify the appropriate dose for future studies by escalating through predetermined dose levels until reaching the maximum tolerated dose (MTD)âthe highest dose that does not cause unacceptable side effects.
In Group 2 (intermittent dosing), dose escalation proceeded without any dose-limiting toxicities observed, and researchers stopped escalation at 200 mg because this dose achieved plasma concentrations sufficient for target inhibition based on preclinical models 1 8 . This 200 mg dose on an intermittent schedule was therefore identified as the recommended phase 2 dose.
The safety results from the trial were particularly encouraging. With the optimized intermittent dosing schedule (14 days on, 14 days off), no dose-limiting toxicities were observed at any dose level up to 200 mg 1 4 8 . This represented a significant improvement over the continuous dosing approach and demonstrated that voruciclib could be administered safely in this vulnerable patient population.
The most common side effects observed across all patients were generally manageable and included:
| Adverse Event | Frequency (%) | Typical Severity |
|---|---|---|
| Diarrhea | 30% | Mild to moderate |
| Nausea | 25% | Mild to moderate |
| Anemia | 22% | Mild to moderate |
| Fatigue | 22% | Mild to moderate |
| Constipation | 17% | Mild |
| Dizziness | 15% | Mild |
| Dyspnea (shortness of breath) | 15% | Mild to moderate |
These side effects are generally consistent with those observed with other targeted cancer therapies and compare favorably with the toxicity profiles of traditional chemotherapy 1 4 8 .
While the primary focus of a phase 1 trial is safety and dosing, researchers also observed early signals of clinical activity:
| Response Category | Number of Patients | Description |
|---|---|---|
| Morphologic leukemia-free state | 1 | Clearance of leukemic blasts from bone marrow |
| Stable disease | 2 | Disease neither significantly grew nor shrank |
| No response | 18 | Disease progression or lack of meeting response criteria |
Though the response rate was modest in this heavily pretreated population, the demonstration of any clinical activity provides proof of concept that CDK9 inhibition can impact these aggressive cancers. Additionally, the disease stabilization observed in some patients suggests that voruciclib may be slowing cancer growth even when not producing dramatic shrinkage 1 4 8 .
Beyond clinical outcomes, the research team conducted sophisticated laboratory analyses to confirm that voruciclib was indeed hitting its intended targets in patients. These biomarker studies revealed three key pieces of evidence:
These molecular findings are crucial because they verify that voruciclib is working through its intended mechanism, providing strong scientific rationale for further development.
The voruciclib study employed several sophisticated research tools to evaluate both the clinical and molecular effects of the drug:
| Research Tool | Application in This Study | Key Insights Generated |
|---|---|---|
| Pharmacokinetic analysis | Measuring drug concentration in blood over time | Confirmed 200 mg dose achieved levels sufficient for target inhibition |
| RNA sequencing | Analyzing gene expression patterns in patient samples | Revealed downregulation of MYC and NF-κB signaling pathways |
| Phosphoprotein assays | Detecting phosphorylation status of RNA polymerase 2 | Provided direct evidence of CDK9 target engagement |
| MTT cytotoxicity assay | Preclinical assessment of drug combination effects | Demonstrated synergy between voruciclib and venetoclax in lab models |
| Dose-limiting toxicity (DLT) assessment | Monitoring specific severe side effects during first treatment cycle | Guided dose escalation decisions and schedule optimization |
These methodologies were essential not only for establishing the safety and preliminary efficacy of voruciclib but also for confirming that the drug was functioning through its intended molecular mechanismâa critical consideration in targeted therapy development 1 2 4 .
The combination of multiple research approaches provided complementary evidence for voruciclib's mechanism of action and therapeutic potential.
The most promising future direction for voruciclib lies in combination therapy with venetoclax. Preclinical studies have demonstrated strong synergy between these two drugs across multiple AML and B-cell malignancy models 1 4 . The scientific rationale is compelling: while venetoclax blocks Bcl-2, voruciclib reduces Mcl-1 levels, simultaneously targeting two key survival proteins that cancers often use interchangeably to resist treatment.
This dual approach essentially creates a one-two punch against cancer's defense system, potentially preventing the development of resistance that often limits the effectiveness of single-agent targeted therapies. Based on the solid scientific rationale and encouraging preclinical data, clinical trials evaluating this combination are already in development 1 8 .
The success of voruciclib's intermittent dosing schedule in improving safety also provides important insights for future drug development strategies targeting transcriptional regulators. CDK9 inhibition's effect on short-lived oncoproteins represents a promising approach that might extend beyond blood cancers to certain solid tumors.
Furthermore, the demonstration that voruciclib can counteract resistance to venetoclax suggests potential applications in treating earlier lines of therapy, potentially benefiting a broader patient population 1 4 . The drug's additional effect on MYC and NF-κB pathwaysâboth important drivers of many cancer typesâfurther expands its potential utility 4 8 .
Evaluate voruciclib in combination with venetoclax
Identify patients most likely to respond to CDK9 inhibition
Explore efficacy in solid tumors dependent on short-lived oncoproteins
Test voruciclib with other targeted therapies and immunotherapies
The successful completion of this phase 1 dose-escalation study of voruciclib represents more than just another early-stage clinical trialâit demonstrates a scientifically sound approach to overcoming one of the most challenging problems in modern oncology: treatment resistance.
By strategically targeting CDK9 to reduce Mcl-1 production, researchers have developed a potentially promising strategy to counter resistance to venetoclax and other targeted therapies. The establishment of a safe dosing schedule (200 mg on days 1-14 of a 28-day cycle) with manageable side effects paves the way for further development, particularly in combination regimens.
While there is still much work to be doneâincluding larger clinical trials to confirm efficacy and optimize patient selectionâthis research represents an important step forward in the evolving landscape of precision oncology. For patients facing limited options after their blood cancers have resisted conventional treatments, the voruciclib story offers genuine hope that scientists are developing increasingly sophisticated strategies to outmaneuver cancer's adaptive defenses.
As Dr. Alexey V. Danilov, one of the senior authors of the study, noted, these findings "pave the way for evaluation of the combination of voruciclib with venetoclax for patients with previously treated AML" 4 8 . With several such studies already in planning, the future of CDK9 inhibition as a therapeutic strategy appears promising indeed.
Safety and dosing established
Next step in development
Target engagement demonstrated
Potential new treatment option